FIELD OF THE INVENTION
[0001] The present invention relates generally to antibodies specific for Glucocorticoid-induced
TNF receptor (GITR) and uses thereof. More specifically, the invention relates to
humanized antibodies that recognize human GITR and modulate its activity, particularly
in immune and proliferative disorders.
BACKGROUND OF THE INVENTION
[0002] Glucocorticoid-induced TNFR-related protein (GITR), a member of the TNFR superfamily,
is expressed in many components of the innate and adaptive immune system (see, e.g.,
Hanabuchi et al. (2006) Blood 107:3617-3623; and
Nocentini and Riccardi (2005) Eur. J. Immunol. 2005. 35:1016-1022). Its membrane expression is increased following T cell activation (Hanabuchi,
supra; and Nocentini and Riccardi,
supra); its triggering co-activates effector T lymphocytes and modulates regulatory T cell
(Treg) activity (see, e.g.,
McHugh, et al. (2002) Immunity 2002. 16:311-323;
Shimizu, et al. (2002) Nat. Immunol.. 3:135-142;
Ronchetti, et al (2004) Eur. J. Immunol. 34:613-622; and
Tone, et al. (2003) Proc. Natl. Acad. Sci. USA 100:15059-15064.
[0005] The need exists for improved methods and compositions for the treatment of immune
and proliferative disorders, e.g., tumors and cancers, by use of agents that modulate
GITR activity. Preferably, such agonists would have a high affinity for the target
molecule, and would be able to stimulate GITR signaling at relatively low doses. Preferably,
such methods and compositions would be highly specific for GITR, and not interfere
with the activity of other receptors. Preferably, such methods and compositions would
employ agonists suitable for modification for the delivery of cytotoxic payloads to
target cells, but also suitable for non-cytotoxic uses. Preferably, such methods and
compositions would employ antibodies modified to limit their antigenicity when administered
to a subject in need thereof.
BRIEF DESCRIPTION OF THE DRAWING
SUMMARY OF THE INVENTION
[0008] The present invention meets these needs in the art and more by providing agonists
of GITR, e.g. humanized anti-GITR antibodies.
[0009] In one aspect the invention provides binding compounds, such as an antibodies or
fragment thereof, including humanized or chimeric recombinant antibodies, that binds
human GITR, comprising an antibody light chain variable domain, or antigen binding
fragment thereof, having at least one or more CDRs selected from the group consisting
of SEQ ID NOs: 56-88 and a heavy chain variable domain, having at least one or more
CDRs selected from the group consisting of SEQ ID NOs: 23-55.
[0010] In other embodiments the binding compound of the present invention comprises a light
chain variable domain and a heavy chain variable domain, or the antigen binding fragments
thereof, described in the preceding two paragraphs.
[0011] In some embodiments, the binding compound comprises a framework region, wherein the
amino acid sequence of the framework region is all or substantially all of a human
immunoglobulin amino acid sequence.
[0012] In some embodiments the light chain variable domain comprises a sequence selected
from the group consisting of SEQ ID NOs: 12-22 or a variant thereof. In some embodiments
the heavy chain variable domain comprises a sequence selected from the group consisting
of SEQ ID NOs: 1-11. In yet a further embodiment, the binding compound comprises a
light chain variable domain and a heavy chain variable domain, or the antigen binding
fragments thereof, described in this paragraph.
[0013] In other embodiments the binding compound of the present invention comprises a light
chain variable domain, or an antigen binding fragment thereof, consisting essentially
of a sequence selected from the group consisting of SEQ ID NOs: 91, 93, 95, 97, 99,
101, 103, 105, 107, 109, 111, and/or a heavy chain variable domain, or an antigen
binding fragment thereof, consisting essentially of a sequence selected from the group
consisting of SEQ ID NOs: 90, 92, 94, 96, 98, 100, 102, 104, 106, 108, 110.
[0014] In one embodiment, the invention relates to antibodies that are able to block the
binding of a binding compound of the present invention to human GITR in a cross-blocking
assay. In various embodiments the antibody is able to block binding of human GITR
to an antibody comprising the CDR sequences of antibodies 36E5, 3D6, 61G6, 6H6, 61F6,
1D8, 17F10, 35D8, 49A1, 9E5, or 31H6 as disclosed herein. In another embodiment, the
invention relates to binding compounds that are able to block GITR-mediated activity,
such activities including but not limited to, co-stimulation of naive CD4+ T cell
proliferation assay.
[0015] In some embodiments, the binding compound of the present invention further comprises
a heavy chain constant region, wherein the heavy chain constant region comprises a
γ1, γ2, γ3, or γ4 human heavy chain constant region or a variant thereof. In various
embodiments the light chain constant region comprises a lambda or a kappa human light
chain constant region.
[0016] In various embodiments the binding compounds of the present invention are polyclonal,
monoclonal, chimeric, humanized or fully human antibodies or fragments thereof. The
present invention also contemplates that the antigen binding fragment is an antibody
fragment selected from the group consisting of Fab, Fab', Fab'-SH, Fv, scFv, F(ab')
2, and a diabody.
[0017] The present invention encompasses a method of enhancing an immune response in a human
subject comprising administering to a subject in need thereof an antibody (or a antigen
binding fragment thereof) specific for GITR in an amount effective to stimulate GITR
signaling. In some embodiments, the antibody specific for GITR is the humanized or
chimeric antibody. In further embodiments, the immune response is an anti-infective
or anti-viral response. In certain embodiments, the GITR antibody or antigen binding
fragment thereof is co-administered with a TGFβ antibody or local radiation.
[0018] The present invention encompasses an isolated nucleic acid encoding the polypeptide
sequence of an antibody embodiment of the binding compound of the present invention.
The nucleic acid can be in an expression vector operably linked to control sequences
recognized by a host cell transfected with the vector. Also encompassed is a host
cell comprising the vector, and a method of producing a polypeptide comprising culturing
the host cell under conditions wherein the nucleic acid sequence is expressed, thereby
producing the polypeptide, and recovering the polypeptide from the host cell or medium.
[0019] The present invention provides an antibody or antigen binding fragment thereof, produced
by a hybridoma deposited at the American Type Culture Collection (ATCC), wherein the
hybridoma is selected from the group consisting of PTA-9889, PTA-9890, PTA-9891, PTA-9892,
PTA-9893, PTA-10286, PTA-10287, PTA-10288, PTA-10289, PTA-10290, and PTA-10291.
[0020] The present invention encompasses an antibody or antigen binding fragment that binds
to human GITR protein, wherein the antibody or antigen binging fragment recognizes
an epitope spanning module 3 and module 4 of human GITR protein (SEQ ID NO: 89). In
certain embodiments, the epitope comprises Gly
57, Arg
65, His
67, Lys
80, Phe
81, Ser
82, and Gln
86. In yet other embodiments the antibody cross-blocks at least one of the antibodies
or antibody fragments produced by the hybridomas selected from group consisting of
PTA-9889, PTA-9890, PTA-9891, PTA-9892, PTA-9893, PTA-10286, PTA-10287, PTA-10288,
PTA-10289, PTA-10290, and PTA-10291.
DETAILED DESCRIPTION
[0021] As used herein, including the appended claims, the singular forms of words such as
"a," "an," and "the," include their corresponding plural references unless the context
clearly dictates otherwise. Table 15 below provides a listing of sequence identifiers
used in this application. All references cited herein are incorporated by reference
to the same extent as if each individual publication, database entry (e.g. Genbank
sequences or GeneID entries), patent application, or patent, was specifically and
individually indicated to be incorporated by reference. Citation of the references
herein is not intended as an admission that any of the foregoing is pertinent prior
art, nor does it constitute any admission as to the contents or date of these publications
or documents.
I. Definitions
[0022] The terms "GITR", "Glucocorticoid-induced TNFR-related protein", "Activation-inducible
TNFR family receptor", "AITR", "Tumor necrosis factor receptor superfamily member
18", and "TNFSF18" are well known in the art. The human and mouse GITR nucleotide
and polypeptide sequences are disclosed in
WO 98/06842. GenBank
® deposits of the human GITR amino sequence (Q9Y5U5) and mouse GITR nucleic and amino
acid sequences (AF109216) are also available.
[0023] "Proliferative activity" encompasses an activity that promotes, that is necessary
for, or that is specifically associated with, e.g., normal cell division, as well
as cancer, tumors, dysplasia, cell transformation, metastasis, and angiogenesis.
[0024] "Administration" and "treatment," as it applies to an animal, human, experimental
subject, cell, tissue, organ, or biological fluid, refers to contact of an exogenous
pharmaceutical, therapeutic, diagnostic agent, or composition to the animal, human,
subject, cell, tissue, organ, or biological fluid. "Administration" and "treatment"
can refer, e.g., to therapeutic, pharmacokinetic, diagnostic, research, and experimental
methods. Treatment of a cell encompasses contact of a reagent to the cell, as well
as contact of a reagent to a fluid, where the fluid is in contact with the cell. "Administration"
and "treatment" also means
in vitro and
ex vivo treatments, e.g., of a cell, by a reagent, diagnostic, binding composition, or by
another cell. "Treatment," as it applies to a human, veterinary, or research subject,
refers to therapeutic treatment, prophylactic or preventative measures, to research
and diagnostic applications. "Treatment" as it applies to a human, veterinary, or
research subject, or cell, tissue, or organ, encompasses contact of an agent with
animal subject, a cell, tissue, physiological compartment, or physiological fluid.
"Treatment of a cell" also encompasses situations where the agent contacts GITR, e.g.,
in the fluid phase or colloidal phase, but also situations where the agonist or antagonist
does not contact the cell or the receptor.
[0025] As used herein, the term "antibody" refers to any form of antibody that exhibits
the desired biological activity. Thus, it is used in the broadest sense and specifically
covers monoclonal antibodies (including full length monoclonal antibodies), polyclonal
antibodies, multispecific antibodies (e.g., bispecific antibodies), chimeric antibodies,
humanized antibodies, fully human antibodies, etc. so long as they exhibit the desired
biological activity.
[0026] As used herein, the terms "GITR binding fragment," "binding fragment thereof" or
"antigen binding fragment thereof" encompass a fragment or a derivative of an antibody
that still substantially retains its biological activity of inducing GITR signaling
referred to herein as "GITR inducing activity." The term "antibody fragment" or GITR
binding fragment refers to a portion of a full length antibody, generally the antigen
binding or variable region thereof. Examples of antibody fragments include Fab, Fab',
F(ab')
2, and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules,
e.g., sc-Fv; and multispecific antibodies formed from antibody fragments. Typically,
a binding fragment or derivative retains at least 10% of its GITR agonist activity.
Preferably, a binding fragment or derivative retains at least 25%, 50%, 60%, 70%,
80%, 90%, 95%, 99% or 100% (or more) of its GITR agonist activity, although any binding
fragment with sufficient affinity to exert the desired biological effect will be useful.
It is also intended that a GITR binding fragment can include variants having conservative
amino acid substitutions that do not substantially alter its biologic activity.
[0027] The term "monoclonal antibody", as used herein, refers to an antibody obtained from
a population of substantially homogeneous antibodies,
i.e., the individual antibodies comprising the population are identical except for possible
naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies
are highly specific, being directed against a single antigenic epitope. In contrast,
conventional (polyclonal) antibody preparations typically include a multitude of antibodies
directed against (or specific for) different epitopes. The modifier "monoclonal" indicates
the character of the antibody as being obtained from a substantially homogeneous population
of antibodies, and is not to be construed as requiring production of the antibody
by any particular method. For example, the monoclonal antibodies to be used in accordance
with the present invention may be made by the hybridoma method first described by
Kohler et al. (1975) Nature 256: 495, or may be made by recombinant DNA methods (
see,
e.g., U.S. Pat. No. 4,816,567). The "monoclonal antibodies" may also be isolated from phage antibody libraries
using the techniques described in
Clackson et al. (1991) Nature 352: 624-628 and
Marks et al. (1991) J. Mol. Biol. 222: 581-597, for example.
[0028] The monoclonal antibodies herein specifically include "chimeric" antibodies (immunoglobulins)
in which a portion of the heavy and/or light chain is identical with or homologous
to corresponding sequences in antibodies derived from a particular species or belonging
to a particular antibody class or subclass, while the remainder of the chain(s) is
identical with or homologous to corresponding sequences in antibodies derived from
another species or belonging to another antibody class or subclass, as well as fragments
of such antibodies, so long as they exhibit the desired biological activity.
U.S. Pat. No. 4,816,567;
Morrison et al. (1984) Proc. Natl. Acad. Sci. USA 81: 6851-6855.
[0029] A "domain antibody" is an immunologically functional immunoglobulin fragment containing
only the variable region of a heavy chain or the variable region of a light chain.
In some instances, two or more V
H regions are covalently joined with a peptide linker to create a bivalent domain antibody.
The two V
H regions of a bivalent domain antibody may target the same or different antigens.
[0030] A "bivalent antibody" comprises two antigen binding sites. In some instances, the
two binding sites have the same antigen specificities. However, bivalent antibodies
may be bispecific (see below).
[0033] As used herein, the term "diabodies" refers to small antibody fragments with two
antigen-binding sites, which fragments comprise a heavy chain variable domain (V
H) connected to a light chain variable domain (V
L) in the same polypeptide chain (V
H-V
L or V
L-V
H). By using a linker that is too short to allow pairing between the two domains on
the same chain, the domains are forced to pair with the complementary domains of another
chain and create two antigen-binding sites. Diabodies are described more fully in,
e.g., EP 404,097;
WO 93/11161; and
Holliger et al. (1993) Proc. Natl. Acad. Sci. USA 90: 6444-6448. For a review of engineered antibody variants generally see
Holliger and Hudson (2005) Nat. Biotechnol. 23:1126-1136.
[0034] As used herein, the term "humanized antibody" refers to forms of antibodies that
contain sequences from non-human (e.g., murine) antibodies as well as human antibodies.
Such antibodies contain minimal sequence derived from non-human immunoglobulin. In
general, the humanized antibody will comprise substantially all of at least one, and
typically two, variable domains, in which all or substantially all of the hypervariable
loops correspond to those of a non-human immunoglobulin and all or substantially all
of the FR regions are those of a human immunoglobulin sequence. The humanized antibody
optionally also will comprise at least a portion of an immunoglobulin constant region
(Fc), typically that of a human immunoglobulin. The prefix "hum", "hu" or "h" is added
to antibody clone designations when necessary to distinguish humanized antibodies
from parental rodent antibodies. The humanized forms of rodent antibodies will generally
comprise the same CDR sequences of the parental rodent antibodies, although certain
amino acid substitutions may be included to increase affinity, increase stability
of the humanized antibody, or for other reasons.
[0035] The antibodies of the present invention also include antibodies with modified (or
blocked) Fc regions to provide altered effector functions. See, e.g.,
U.S. Pat. No. 5,624,821;
WO2003/086310;
WO2005/120571;
WO2006/0057702;
Presta (2006) Adv. Drug Delivery Rev. 58:640-656. Such modification can be used to enhance or suppress various reactions of the immune
system, with possible beneficial effects in diagnosis and therapy. Alterations of
the Fc region include amino acid changes (substitutions, deletions and insertions),
glycosylation or deglycosylation, and adding multiple Fc. Changes to the Fc can also
alter the half-life of antibodies in therapeutic antibodies, and a longer half-life
would result in less frequent dosing, with the concomitant increased convenience and
decreased use of material.
See Presta (2005) J. Allergy Clin. Immunol. 116:731 at 734-35.
[0036] The antibodies of the present invention also include antibodies with intact Fc regions
that provide full effector functions, e.g. antibodies of isotype IgG1, which induce
complement-dependent cytotoxicity (CDC) or antibody dependent cellular cytotoxicity
(ADCC) in a targeted cell.
[0037] The antibodies of the present invention also include antibodies conjugated to cytotoxic
payloads, such as cytotoxic agents or radionuclides. Such antibody conjugates may
be used in immunotherapy in conjunction with anti-GITR treatment, to selectively target
and kill cells expressing certain antigens on their surface. Exemplary cytotoxic agents
include ricin, vinca alkaloid, methotrexate,
Psuedomonas exotoxin, saporin, diphtheria toxin, cisplatin, doxorubicin, abrin toxin, gelonin
and pokeweed antiviral protein. Exemplary radionuclides for use in immunotherapy with
the antibodies of the present invention include
121I,
131I,
90Y,
67Cu,
211At,
177Lu,
143Pr and
213Bi.
See, e.g., U.S. Patent Application Publication No. 2006/0014225.
[0038] The term "fully human antibody" refers to an antibody that comprises human immunoglobulin
protein sequences only. A fully human antibody may contain murine carbohydrate chains
if produced in a mouse, in a mouse cell, or in a hybridoma derived from a mouse cell.
Similarly, "mouse antibody" or "rat antibody" refer to an antibody that comprises
only mouse or rat immunoglobulin sequences, respectively. A fully human antibody may
be generated in a human being, in a transgenic animal having human immunoglobulin
germline sequences, by phage display or other molecular biological methods.
[0039] As used herein, the term "hypervariable region" refers to the amino acid residues
of an antibody that are responsible for antigen-binding. The hypervariable region
comprises amino acid residues from a "complementarity determining region" or "CDR"
(
e.g. residues 24-34 (CDRL1), 50-56 (CDRL2) and 89-97 (CDRL3) in the light chain variable
domain and residues 31-35 (CDRH1), 50-65 (CDRH2) and 95-102 (CDRH3) in the heavy chain
variable domain (Kabat
et al. (1991) Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service,
National Institutes of Health, Bethesda, Md.) and/or those residues from a "hypervariable
loop" (
e.g.,. residues 26-32 (L1), 50-52 (L2) and 91-96 (L3) in the light chain variable domain
and 26-32 (H1), 53-55 (H2) and 96-101 (H3) in the heavy chain variable domain (
Chothia and Lesk (1987) J. Mol. Biol. 196: 901-917). As used herein, the term "framework" or "FR" residues refers to those variable
domain residues other than the hypervariable region residues defined herein as CDR
residues. The residue numbering above relates to the Kabat numbering system and does
not necessarily correspond in detail to the sequence numbering in the accompanying
Sequence Listing.
[0040] "Binding compound" refers to a molecule, small molecule, macromolecule, polypeptide,
antibody or fragment or analogue thereof, or soluble receptor, capable of binding
to a target. "Binding compound" also may refer to a complex of molecules, e.g., a
non-covalent complex, to an ionized molecule, and to a covalently or non-covalently
modified molecule, e.g., modified by phosphorylation, acylation, cross-linking, cyclization,
or limited cleavage, that is capable of binding to a target. When used with reference
to antibodies, the term "binding compound" refers to both antibodies and antigen binding
fragments thereof. "Binding" refers to an association of the binding composition with
a target where the association results in reduction in the normal Brownian motion
of the binding composition, in cases where the binding composition can be dissolved
or suspended in solution. "Binding composition" refers to a molecule, e.g. a binding
compound, in combination with a stabilizer, excipient, salt, buffer, solvent, or additive,
capable of binding to a target.
[0041] "Conservatively modified variants" or "conservative substitution" refers to substitutions
of amino acids are known to those of skill in this art and may often be made even
in essential regions of the polypeptide without altering the biological activity of
the resulting molecule. Such exemplary substitutions are preferably made in accordance
with those set forth in Table 1 as follows:
Table 1
| Exemplary Conservative Amino Acid Substitutions |
| Original residue |
Conservative substitution |
| Ala (A) |
Gly; Ser |
| Arg (R) |
Lys, His |
| Asn (N) |
Gin; His |
| Asp (D) |
Glu; Asn |
| Cys (C) |
Ser; Ala |
| Gln (Q) |
Asn |
| Glu (E) |
Asp; Gin |
| Gly (G) |
Ala |
| His (H) |
Asn; Gin |
| Ile (I) |
Leu; Val |
| Leu (L) |
Ile; Val |
| Lys (K) |
Arg; His |
| Met (M) |
Leu; Ile; Tyr |
| Phe (F) |
Tyr; Met; Leu |
| Pro (P) |
Ala |
| Ser (S) |
Thr |
| Thr (T) |
Ser |
| Trp (W) |
Tyr; Phe |
| Tyr (Y) |
Trp; Phe |
| Val (V) |
Ile; Leu |
[0043] The phrase "consists essentially of," or variations such as "consist essentially
of" or "consisting essentially of," as used throughout the specification and claims,
indicate the inclusion of any recited elements or group of elements, and the optional
inclusion of other elements, of similar or different nature than the recited elements,
that do not materially change the basic or novel properties of the specified dosage
regimen, method, or composition. As a non-limiting example, a binding compound that
consists essentially of a recited amino acid sequence may also include one or more
amino acids, including substitutions of one or more amino acid residues, that do not
materially affect the properties of the binding compound.
[0044] "Effective amount" encompasses an amount sufficient to ameliorate or prevent a symptom
or sign of the medical condition. Effective amount also means an amount sufficient
to allow or facilitate diagnosis. An effective amount for a particular patient or
veterinary subject may vary depending on factors such as the condition being treated,
the overall health of the patient, the method route and dose of administration and
the severity of side affects.
See, e.g., U.S. Pat. No. 5,888,530. An effective amount can be the maximal dose or dosing protocol that avoids significant
side effects or toxic effects. The effect will result in an improvement of a diagnostic
measure or parameter by at least 5%, usually by at least 10%, more usually at least
20%, most usually at least 30%, preferably at least 40%, more preferably at least
50%, most preferably at least 60%, ideally at least 70%, more ideally at least 80%,
and most ideally at least 90%, where 100% is defined as the diagnostic parameter shown
by a normal subject.
See, e.g., Maynard et al. (1996) A Handbook of SOPs for Good Clinical Practice, Interpharm Press,
Boca Raton, FL;
Dent (2001) Good Laboratory and Good Clinical Practice, Urch Publ., London, UK.
[0045] "Immune condition" or "immune disorder" encompasses, e.g., pathological inflammation,
an inflammatory disorder, and an autoimmune disorder or disease. "Immune condition"
also refers to infections, persistent infections, and proliferative conditions, such
as cancer, tumors, and angiogenesis, including infections, tumors, and cancers that
resist eradication by the immune system. "Cancerous condition" includes, e.g., cancer,
cancer cells, tumors, angiogenesis, and precancerous conditions such as dysplasia.
[0046] The term immune disorder means a disease in which a component of the immune system
of a mammal causes, mediates or otherwise contributes to a morbidity in the mammal.
Also included are diseases in which stimulation or intervention of the immune response
has an ameliorative effect on progression of the disease. Included within this term
are autoimmune diseases, immune-mediated inflammatory diseases, non-immune-mediated
inflammatory diseases, infectious diseases, and immunodeficiency diseases. Examples
of immune-related and inflammatory diseases, some of which are immune or T cell mediated,
which can be treated according to the invention include systemic lupus erythematosis,
rheumatoid arthritis, juvenile chronic arthritis, spondyloarthropathies, systemic
sclerosis (scleroderma), idiopathic inflammatory myopathies (dermatomyositis, polymyositis),
Sjogren's syndrome, systemic vasculitis, sarcoidosis, autoimmune hemolytic anemia
(immune pancytopenia, paroxysmal nocturnal hemoglobinuria), autoimmune thrombocytopenia
(idiopathic thrombocytopenic purpura, immune-mediated thrombocytopenia), thyroiditis
(Grave's disease, Hashimoto's thyroiditis, juvenile lymphocytic thyroiditis, atrophic
thyroiditis), diabetes mellitus, immune-mediated renal disease (glomerulonephritis,
tubulointerstitial nephritis), demyelinating diseases of the central and peripheral
nervous systems such as multiple sclerosis, idiopathic demyelinating polyneuropathy
or Guillain-Barre syndrome, and chronic inflammatory demyelinating polyneuropathy,
hepatobiliary diseases such as infectious hepatitis (hepatitis A, B, C, D, E and other
non-hepatotropic viruses), autoimmune chronic active hepatitis, primary biliary cirrhosis,
granulomatous hepatitis, and sclerosing cholangitis, inflammatory and fibrotic lung
diseases such as inflammatory bowel disease (ulcerative colitis: Crohn's disease),
gluten-sensitive enteropathy, and Whipple's disease, autoimmune or immune-mediated
skin diseases including bullous skin diseases, erythema multiforme and contact dermatitis,
psoriasis, allergic diseases such as asthma, allergic rhinitis, atopic dermatitis,
food hypersensitivity and urticaria, immunologic diseases of the lung such as eosinophilic
pneumonias, idiopathic pulmonary fibrosis and hypersensitivity pneumonitis, transplantation
associated diseases including graft rejection and graft-versus-host-disease. Infectious
diseases include AIDS (HIV infection), hepatitis A, B, C, D, and E, bacterial infections,
fungal infections, protozoal infections and parasitic infections.
[0047] The terms "cancer", "tumor", "cancerous", and "malignant" refer to or describe the
physiological condition in mammals that is typically characterized by unregulated
cell growth. Examples of cancer include but are not limited to, carcinoma including
adenocarcinoma, lymphoma, blastoma, melanoma, sarcoma, and leukemia. More particular
examples of such cancers include squamous cell cancer, small-cell lung cancer, non-small
cell lung cancer, gastrointestinal cancer, Hodgkin's and non-Hodgkin's lymphoma, pancreatic
cancer, glioblastoma, glioma, cervical cancer, ovarian cancer, liver cancer such as
hepatic carcinoma and hepatoma, bladder cancer, breast cancer, colon cancer, colorectal
cancer, endometrial carcinoma, myeloma (such as multiple myeloma), salivary gland
carcinoma, kidney cancer such as renal cell carcinoma and Wilms' tumors, basal cell
carcinoma, melanoma, prostate cancer, vulval cancer, thyroid cancer, testicular cancer,
esophageal cancer, and various types of head and neck cancer.
[0048] As cancerous cells grow and multiply, they form a mass of cancerous tissue, that
is a tumor, which invades and destroys normal adjacent tissues. Malignant tumors are
cancer. Malignant tumors usually can be removed, but they may grow back. Cells from
malignant tumors can invade and damage nearby tissues and organs. Also, cancer cells
can break away from a malignant tumor and enter the bloodstream or lymphatic system,
which is the way cancer cells spread from the primary tumor (
i.e., the original cancer) to form new tumors in other organs. The spread of cancer in
the body is called metastasis (
What You Need to Know About Cancer- an Overview, NIH Publication No. 00-1566; posted
Sept. 26, 2000, updated Sept. 16, 2002 (2002)).
[0049] As used herein, the term "solid tumor" refers to an abnormal growth or mass of tissue
that usually does not contain cysts or liquid areas. Solid tumors may be benign (not
cancerous) or malignant (cancerous). Different types of solid tumors are named for
the type of cells that form them. Examples of solid tumors are sarcomas, carcinomas,
and lymphomas. Leukemias (cancers of the blood) generally do not form solid tumors
(National Cancer Institute, Dictionary of Cancer Terms).
[0051] As used herein, the term "carcinoma
in situ" refers to cancerous cells that are still contained within the tissue where they
started to grow, and have not yet become invasive or spread to other parts of the
body.
[0052] As used herein, the term "carcinomas" refers to cancers of epithelial cells, which
are cells that cover the surface of the body, produce hormones, and make up glands.
Examples of carcinomas are cancers of the skin, lung, colon, stomach, breast, prostate
and thyroid gland.
[0053] As used herein, the term "isolated nucleic acid molecule" refers to a nucleic acid
molecule that is identified and separated from at least one contaminant nucleic acid
molecule with which it is ordinarily associated in the natural source of the antibody
nucleic acid. An isolated nucleic acid molecule is other than in the form or setting
in which it is found in nature. Isolated nucleic acid molecules therefore are distinguished
from the nucleic acid molecule as it exists in natural cells. However, an isolated
nucleic acid molecule includes a nucleic acid molecule contained in cells that ordinarily
express the antibody where, for example, the nucleic acid molecule is in a chromosomal
location different from that of natural cells.
[0054] The expression "control sequences" refers to DNA sequences involved in the expression
of an operably linked coding sequence in a particular host organism. The control sequences
that are suitable for prokaryotes, for example, include a promoter, optionally an
operator sequence, and a ribosome binding site. Eukaryotic cells are known to use
promoters, polyadenylation signals, and enhancers.
[0055] A nucleic acid is "operably linked" when it is placed into a functional relationship
with another nucleic acid sequence. For example, DNA for a presequence or secretory
leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein
that participates in the secretion of the polypeptide; a promoter or enhancer is operably
linked to a coding sequence if it affects the transcription of the sequence; or a
ribosome binding site is operably linked to a coding sequence if it is positioned
so as to facilitate translation. Generally, "operably linked" means that the DNA sequences
being linked are contiguous, and, in the case of a secretory leader, contiguous and
in reading frame. However, enhancers do not have to be contiguous. Linking is accomplished
by ligation at convenient restriction sites. If such sites do not exist, the synthetic
oligonucleotide adaptors or linkers are used in accordance with conventional practice.
[0056] As used herein, the expressions "cell," "cell line," and "cell culture" are used
interchangeably and all such designations include progeny. Thus, the words "transformants"
and "transformed cells" include the primary subject cell and cultures derived therefrom
without regard for the number of transfers. It is also understood that all progeny
may not be precisely identical in DNA content, due to deliberate or inadvertent mutations.
Mutant progeny that have the same function or biological activity as screened for
in the originally transformed cell are included. Where distinct designations are intended,
it will be clear from the context.
[0057] As used herein, "polymerase chain reaction" or "PCR" refers to a procedure or technique
in which minute amounts of a specific piece of nucleic acid, RNA and/or DNA, are amplified
as described in,
e.g., U.S. Pat. No. 4,683,195. Generally, sequence information from the ends of the region of interest or beyond
needs to be available, such that oligonucleotide primers can be designed; these primers
will be identical or similar in sequence to opposite strands of the template to be
amplified. The 5' terminal nucleotides of the two primers can coincide with the ends
of the amplified material. PCR can be used to amplify specific RNA sequences, specific
DNA sequences from total genomic DNA, and cDNA transcribed from total cellular RNA,
bacteriophage or plasmid sequences, etc.
See generally Mullis et al. (1987) Cold Spring Harbor Symp. Quant. Biol. 51:263;
Erlich, ed., (1989) PCR TECHNOLOGY (Stockton Press, N.Y.) As used herein, PCR is considered to be one, but not the only, example of a nucleic
acid polymerase reaction method for amplifying a nucleic acid test sample comprising
the use of a known nucleic acid as a primer and a nucleic acid polymerase to amplify
or generate a specific piece of nucleic acid.
[0058] As used herein, the term "germline sequence" refers to a sequence of unrearranged
immunoglobulin DNA sequences, including rodent (e.g. mouse) and human germline sequences.
Any suitable source of unrearranged immunoglobulin DNA may be used. Human germline
sequences may be obtained, for example, from JOINSOLVER
® germline databases on the website for the National Institute of Arthritis and Musculoskeletal
and Skin Diseases of the United States National Institutes of Health. Mouse germline
sequences may be obtained, for example, as described in
Giudicelli et al. (2005) Nucleic Acids Res. 33:D256-D261.
[0059] To examine the extent of enhancement of GITR activity, for example, samples or assays
comprising a given, e.g., protein, gene, cell, or organism, are treated with a potential
activating or inhibiting agent and are compared to control samples without the agent.
Control samples, i.e., not treated with agent, are assigned a relative activity value
of 100%. Inhibition is achieved when the activity value relative to the control is
about 90% or less, typically 85% or less, more typically 80% or less, most typically
75% or less, generally 70% or less, more generally 65% or less, most generally 60%
or less, typically 55% or less, usually 50% or less, more usually 45% or less, most
usually 40% or less, preferably 35% or less, more preferably 30% or less, still more
preferably 25% or less, and most preferably less than 20%. Activation is achieved
when the activity value relative to the control is about 110%, generally at least
120%, more generally at least 140%, more generally at least 160%, often at least 180%,
more often at least 2-fold, most often at least 2.5-fold, usually at least 5-fold,
more usually at least 10-fold, preferably at least 20-fold, more preferably at least
40-fold, and most preferably over 40-fold higher.
[0060] Endpoints in activation or inhibition can be monitored as follows. Activation, inhibition,
and response to treatment, e.g., of a cell, physiological fluid, tissue, organ, and
animal or human subject, can be monitored by an endpoint. The endpoint may comprise
a predetermined quantity or percentage of, e.g., an indicia of inflammation, oncogenicity,
or cell degranulation or secretion, such as the release of a cytokine, toxic oxygen,
or a protease. The endpoint may comprise, e.g., a predetermined quantity of ion flux
or transport; cell migration; cell adhesion; cell proliferation; potential for metastasis;
cell differentiation; and change in phenotype, e.g., change in expression of gene
relating to inflammation, apoptosis, transformation, cell cycle, or metastasis (see,
e.g.,
Knight (2000) Ann. Clin. Lab. Sci. 30:145-158;
Hood and Cheresh (2002) Nature Rev. Cancer 2:91-100;
Timme et al. (2003) Curr. Drug Targets 4:251-261;
Robbins and Itzkowitz (2002) Med. Clin. North Am. 86:1467-1495;
Grady and Markowitz (2002) Annul. Rev. Genomics Hum. Genet. 3:101-128;
Bauer, et al. (2001) Glia 36:235-243;
Stanimirovic and Satoh (2000) Brain Pathol. 10:113-126).
[0061] An endpoint of inhibition is generally 75% of the control or less, preferably 50%
of the control or less, more preferably 25% of the control or less, and most preferably
10% of the control or less. Generally, an endpoint of activation is at least 150%
the control, preferably at least two times the control, more preferably at least four
times the control, and most preferably at least 10 times the control.
[0062] "Small molecule" is defined as a molecule with a molecular weight that is less than
10 kDa, typically less than 2 kDa, and preferably less than 1 kDa. Small molecules
include, but are not limited to, inorganic molecules, organic molecules, organic molecules
containing an inorganic component, molecules comprising a radioactive atom, synthetic
molecules, peptide mimetics, and antibody mimetics. As a therapeutic, a small molecule
may be more permeable to cells, less susceptible to degradation, and less apt to elicit
an immune response than large molecules. Small molecules, such as peptide mimetics
of antibodies and cytokines, as well as small molecule toxins are described.
See, e.g., Casset et al. (2003) Biochem. Biophys. Res. Commun. 307:198-205;
Muyldermans (2001) J. Biotechnol. 74:277-302;
Li (2000) Nat. Biotechnol. 18:1251-1256;
Apostolopoulos et al. (2002) Curr. Med. Chem. 9:411-420;
Monfardini et al. (2002) Curr. Pharm. Des. 8:2185-2199;
Domingues et al. (1999) Nat. Struct. Biol. 6:652-656;
Sato and Sone (2003) Biochem. J. 371:603-608;
U.S. Patent No. 6,326,482.
[0063] "Specifically" or "selectively" binds, when referring to a ligand/receptor, antibody/antigen,
or other binding pair, indicates a binding reaction that is determinative of the presence
of the protein in a heterogeneous population of proteins and other biologics. Thus,
under designated conditions, a specified ligand binds to a particular receptor and
does not bind in a significant amount to other proteins present in the sample. As
used herein, an antibody is said to bind specifically to a polypeptide
comprising a given sequence (in this case GITR) if it binds to polypeptides comprising the sequence
of GITR but does not bind to proteins lacking the sequence of GITR. For example, an
antibody that specifically binds to a polypeptide comprising GITR may bind to a FLAG
®-tagged form of GITR but will not bind to other FLAG
®-tagged proteins.
[0064] The antibody, or binding composition derived from the antigen-binding site of an
antibody, of the contemplated method binds to its antigen with an affinity that is
at least two fold greater, preferably at least ten times greater, more preferably
at least 20-times greater, and most preferably at least 100-times greater than the
affinity with unrelated antigens. In a preferred embodiment the antibody will have
an affinity that is greater than about 10
9 liters/mol, as determined, e.g., by Scatchard analysis.
Munsen et al. (1980) Analyt. Biochem. 107:220-239.
[0065] "Chronic viral infection" or "persistent viral infection" as used herein, is meant
a viral infection of humans or other animals which is able to infect a host and reproduce
within the cells of a host over a prolonged period of time--usually weeks, months
or years, without proving fatal. Amongst viruses giving rise to chronic infections
and which may be treated in accordance with the present invention are the human papilloma
viruses (HPV), Herpes simplex and other herpes viruses, the viruses of hepatitis B
and C (HBV and HCV) as well as other hepatitis viruses, the measles virus, all of
which can produce important clinical diseases, and HIV. Prolonged infection may ultimately
lead to the induction of disease which may be, e. g. in the case of hepatitis C virus
liver cancer, fatal to the patient. Other chronic viral infections which may be treated
in accordance with the present invention include Epstein Barr virus (EBV), as well
as other viruses such as those which may be associated with tumors, or in the case
of animals, various veterinary viral diseases, for example those of domestic pets
or farmyard animals important in agriculture.
[0066] The term "antiviral activity" refers to an inhibition of viral transmission to uninfected
cells, inhibition of the replication of a virus, prevention of the virus from establishing
itself in a host, or ameliorating or alleviating the symptoms of the disease caused
by viral infection. These effects can be evidenced by a reduction in viral load or
decrease in mortality and/or morbidity, which assays are described infra. An antiviral
agent or drug, has antiviral activity and is useful for treating persistent or chronic
viral infections alone, or as part of a multi-drug combination therapy.
II. General
[0067] The present invention provides engineered anti-GITR antibodies and uses thereof to
treat immune disorders, in particular impaired response to infectious diseases (including
viral infections) and cancer.
[0068] GITR, also known as TNFRSF18, is a receptor belonging to the TNR-R superfamily. To
date, crystal structures of human or mouse GITR are not available, however, a modular
architecuture of the molecule, based upon studies described, e.g., in
Naismith and Sprang (1998) Trends Biochem. Sci. 23:74-79, can be established. Figure 2 illustrates that human GITR can be divided into 6 modules.
From the studies below, certain antibodies having agonist activity may have conformational
epitopes that span modules 3 and 4.
II. Generation of GITR Specific Antibodies
[0069] Any suitable method for generating monoclonal antibodies may be used. For example,
a recipient may be immunized with GITR or a fragment thereof. Any suitable method
of immunization can be used. Such methods can include adjuvants, other immunostimulants,
repeated booster immunizations, and the use of one or more immunization routes. Any
suitable source of GITR can be used as the immunogen for the generation of the non-human
antibody of the compositions and methods disclosed herein. Such forms include, but
are not limited whole protein, peptide(s), and epitopes generated through recombinant,
synthetic, chemical or enzymatic degradation means known in the art. In preferred
embodiments the immunogen comprises the extracellular portion of GITR.
[0070] Any form of the antigen can be used to generate the antibody that is sufficient to
generate a biologically active antibody. Thus, the eliciting antigen may be a single
epitope, multiple epitopes, or the entire protein alone or in combination with one
or more immunogenicity enhancing agents known in the art. The eliciting antigen may
be an isolated full-length protein, a cell surface protein (e.g., immunizing with
cells transfected with at least a portion of the antigen), or a soluble protein (e.g.,
immunizing with only the extracellular domain portion of the protein). The antigen
may be produced in a genetically modified cell. The DNA encoding the antigen may genomic
or non-genomic (e.g., cDNA) and encodes at least a portion of the extracellular domain.
As used herein, the term "portion" refers to the minimal number of amino acids or
nucleic acids, as appropriate, to constitute an immunogenic epitope of the antigen
of interest. Any genetic vectors suitable for transformation of the cells of interest
may be employed, including but not limited to adenoviral vectors, plasmids, and non-viral
vectors, such as cationic lipids.
[0071] Any suitable method can be used to elicit an antibody with the desired biologic properties
to enhance GITR signaling. It is desirable to prepare monoclonal antibodies (mAbs)
from various mammalian hosts, such as mice, rats, other rodents, humans, other primates,
etc. Description of techniques for preparing such monoclonal antibodies may be found
in,
e.g., Stites et al. (eds.) BASIC AND CLINICAL IMMUNOLOGY (4th ed.) Lange Medical Publications,
Los Altos, CA, and references cited therein;
Harlow and Lane (1988) ANTIBODIES: A LABORATORY MANUAL CSH Press;
Goding (1986) MONOCLONAL ANTIBODIES: PRINCIPLES AND PRACTICE (2d ed.) Academic Press,
New York, NY. Thus, monoclonal antibodies may be obtained by a variety of techniques familiar
to researchers skilled in the art. Typically, spleen cells from an animal immunized
with a desired antigen are immortalized, commonly by fusion with a myeloma cell.
See Kohler and Milstein (1976) Eur. J. Immunol. 6:511-519. Alternative methods of immortalization include transformation with Epstein Barr
Virus, oncogenes, or retroviruses, or other methods known in the art.
See, e.g., Doyle et al. (eds. 1994 and periodic supplements) CELL AND TISSUE CULTURE: LABORATORY
PROCEDURES, John Wiley and Sons, New York, NY. Colonies arising from single immortalized cells are screened for production of antibodies
of the desired specificity and affinity for the antigen, and yield of the monoclonal
antibodies produced by such cells may be enhanced by various techniques, including
injection into the peritoneal cavity of a vertebrate host. Alternatively, one may
isolate DNA sequences that encode a monoclonal antibody or a antigen binding fragment
thereof by screening a DNA library from human B cells according,
e.g., to the general protocol outlined by
Huse et al. (1989) Science 246:1275-1281.
[0072] Other suitable techniques involve selection of libraries of antibodies in phage or
similar vectors.
See, e.g., Huse
et al. supra; and
Ward et al. (1989) Nature 341:544-546. The polypeptides and antibodies of the present invention may be used with or without
modification, including chimeric or humanized antibodies. Frequently, the polypeptides
and antibodies will be labeled by joining, either covalently or non-covalently, a
substance that provides for a detectable signal. A wide variety of labels and conjugation
techniques are known and are reported extensively in both the scientific and patent
literature. Suitable labels include radionuclides, enzymes, substrates, cofactors,
inhibitors, fluorescent moieties, chemiluminescent moieties, magnetic particles, and
the like. Patents teaching the use of such labels include
U.S. Patent Nos. 3,817,837;
3,850,752;
3,939,350;
3,996,345;
4,277,437;
4,275,149; and
4,366,241. Also, recombinant immunoglobulins may be produced,
see Cabilly U.S. Patent No. 4,816,567; and
Queen et al. (1989) Proc. Nat'l Acad. Sci. USA 86:10029-10033; or made in transgenic mice, see
Mendez et al. (1997) Nature Genetics 15:146-156.
See also Abgenix and Medarex technologies.
[0073] Alternatively, monoclonal antibodies can be produced by enrichment of clonal populations
of B cells isolated from spleens of animals (e.g., mice, rats, rabbits, etc.) immunized
with human GITR (see, e.g.,
WO2008045140,
US5627052, and
US20030186327).
[0074] Antibodies or binding compositions against predetermined fragments of GITR can be
raised by immunization of animals with conjugates of the polypeptide, fragments, peptides,
or epitopes with carrier proteins. Monoclonal antibodies are prepared from cells secreting
the desired antibody. These antibodies can be screened for binding to normal or defective
GITR. These monoclonal antibodies will usually bind with at least a K
d of about 1 µM, more usually at least about 300 nM, 30 nM, 10 nM, 3 nM, 1 nM, 300
pM, 100 pM, 30 pM or better, usually determined by ELISA or Biacore. Suitable non-human
antibodies may also be identified using the biologic assays described in Examples
5 and 6, below.
[0075] Hybridomas corresponding to clones 36E5, 3D6, 61G6, 6H6 and 61F6 were deposited at
the American Type Culture Collection ("ATCC") under the Budapest Treaty requirements,
as PTA-9890, PTA-9889, PTA-9891, PTA-9892, and PTA-9893, respectively, on March 25,
2009.
[0076] Hybridomas corresponding to clones 1D8, 17F10, 35D8, 49A1, 9E5, and 31H6 were deposited
at the ATCC in accordance with the Budapest Treaty requirements on August 21, 2009,
as PTA-10286, PTA-10287, PTA-10288, PTA-10289, PTA-10290, and PTA-10291.
IV. Humanization of GITR Specific Antibodies
[0077] Any suitable non-human antibody can be used as a source for the hypervariable region.
Sources for non-human antibodies include, but are not limited to, murine (e.g.
Mus musculus), rat (e.g.
Rattus norvegicus), Lagomorphs (including rabbits), bovine, and primates. For the most part, humanized
antibodies are human immunoglobulins (recipient antibody) in which hypervariable region
residues of the recipient are replaced by hypervariable region residues from a non-human
species (donor antibody) such as mouse, rat, rabbit or non-human primate having the
desired specificity, affinity, and capacity. In some instances, Fv framework region
(FR) residues of the human immunoglobulin are replaced by corresponding non-human
residues. Furthermore, humanized antibodies may comprise residues that are not found
in the recipient antibody or in the donor antibody. These modifications are made to
further refine antibody performance of the desired biological activity. For further
details, see
Jones et al. (1986) Nature 321:522-525;
Reichmann et al. (1988) Nature 332:323-329; and
Presta (1992) Curr. Op. Struct. Biol. 2:593-596.
[0079] Amino acid sequence variants of humanized anti-GITR antibody are prepared by introducing
appropriate nucleotide changes into the humanized anti-GITR antibody DNA, or by peptide
synthesis. Such variants include, for example, deletions from, and/or insertions into,
and/or substitutions of, residues within the amino acid sequences shown for the humanized
anti-GITR antibody. Any combination of deletion, insertion, and substitution is made
to arrive at the final construct, provided that the final construct possesses the
desired characteristics. The amino acid changes also may alter post-translational
processes of the humanized anti-GITR antibody, such as changing the number or position
of glycosylation sites.
[0080] A useful method for identification of certain residues or regions of the humanized
anti-GITR antibody polypeptide that are preferred locations for mutagenesis is called
"alanine scanning mutagenesis," as described by
Cunningham and Wells (1989) Science 244: 1081-1085. Here, a residue or group of target residues are identified (e.g., charged residues
such as Arg, Asp, His, Lys, and Glu) and replaced by a neutral or negatively charged
amino acid (most preferably alanine or polyalanine) to affect the interaction of the
amino acids with GITR antigen. The amino acid residues demonstrating functional sensitivity
to the substitutions then are refined by introducing further or other variants at,
or for, the sites of substitution. Thus, while the site for introducing an amino acid
sequence variation is predetermined, the nature of the mutation
per se need not be predetermined. For example, to analyze the performance of a mutation
at a given site, Ala scanning or random mutagenesis is conducted at the target codon
or region and the expressed humanized anti-GITR antibody variants are screened for
the desired activity.
[0081] Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging
in length from one residue to polypeptides containing a hundred or more residues,
as well as intrasequence insertions of single or multiple amino acid residues. Examples
of terminal insertions include humanized anti-GITR antibody with an N-terminal methionyl
residue or the antibody fused to an epitope tag. Other insertional variants of the
humanized anti-GITR antibody molecule include the fusion to the N- or C-terminus of
humanized anti-GITR antibody of an enzyme or a polypeptide that increases the serum
half-life of the antibody.
[0082] Another type of variant is an amino acid substitution variant. These variants have
at least one amino acid residue in the humanized anti-GITR antibody molecule removed
and a different residue inserted in its place. The sites of greatest interest for
substitutional mutagenesis include the hypervariable loops, but FR alterations are
also contemplated.
[0083] Another type of amino acid variant of the antibody alters the original glycosylation
pattern of the antibody. By altering is meant deleting one or more carbohydrate moieties
found in the antibody, and/or adding one or more glycosylation sites that are not
present in the antibody. Glycosylation of antibodies is typically either N-linked
or O-linked. N-linked refers to the attachment of the carbohydrate moiety to the side
chain of an asparagine residue. The tripeptide sequences asparagine-X-serine and asparagine-X-threonine,
where X is any amino acid except proline, are the recognition sequences for enzymatic
attachment of the carbohydrate moiety to the asparagine side chain. Thus, the presence
of either of these tripeptide sequences in a polypeptide creates a potential glycosylation
site. O-linked glycosylation refers to the attachment of one of the sugars N-acetylgalactosamine,
galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although
5-hydroxyproline or 5-hydroxylysine may also be used.
[0084] Addition of glycosylation sites to the antibody is conveniently accomplished by altering
the amino acid sequence such that it contains one or more of the above-described tripeptide
sequences (for N-linked glycosylation sites). The alteration may also be made by the
addition of, or substitution by, one or more serine or threonine residues to the sequence
of the original antibody (for O-linked glycosylation sites).
[0085] Yet another type of amino acid variant is the substitution of residues to provide
for greater chemical stability of the final humanized antibody. For example, an asparagine
(N) residue may be changed to reduce the potential for formation of isoaspartate at
any NG sequences within a rodent CDR. A similar problem may occur at a DG sequence.
Reissner and Aswad (2003) Cell. Mol. Life Sci. 60:1281. Isoaspartate formation may debilitate or completely abrogate binding of an antibody
to its target antigen.
Presta (2005) J. Allergy Clin. Immunol. 116:731 at 734. In one embodiment, the asparagine is changed to glutamine (Q). In addition, methionine
residues in rodent CDRs may be changed to reduce the possibility that the methionine
sulfur would oxidize, which could reduce antigen binding affinity and also contribute
to molecular heterogeneity in the final antibody preparation.
Id. In one embodiment, the methionine is changed to alanine (A). Antibodies with such
substitutions are subsequently screened to ensure that the substitutions do not decrease
GITR binding affinity to unacceptable levels.
[0086] Nucleic acid molecules encoding amino acid sequence variants of humanized GITR specific
antibody are prepared by a variety of methods known in the art. These methods include,
but are not limited to, isolation from a natural source (in the case of naturally
occurring amino acid sequence variants) or preparation by oligonucleotide-mediated
(or site-directed) mutagenesis, PCR mutagenesis, and cassette mutagenesis of an earlier
prepared variant or a non-variant version of humanized anti-GITR antibody.
[0087] Ordinarily, amino acid sequence variants of the humanized anti-GITR antibody will
have an amino acid sequence having at least 75% amino acid sequence identity with
the original humanized antibody amino acid sequences of either the heavy or the light
chain more preferably at least 80%, more preferably at least 85%, more preferably
at least 90%, and most preferably at least 95%, 98% or 99%. Identity or homology with
respect to this sequence is defined herein as the percentage of amino acid residues
in the candidate sequence that are identical with the humanized anti-GITR residues,
after aligning the sequences and introducing gaps, if necessary, to achieve the maximum
percent sequence identity, and not considering any conservative substitutions as part
of the sequence identity. None of N-terminal, C-terminal, or internal extensions,
deletions, or insertions into the antibody sequence shall be construed as affecting
sequence identity or homology.
[0088] The humanized antibody can be selected from any class of immunoglobulins, including
IgM, IgG, IgD, IgA, and IgE. Preferably, the antibody is an IgG antibody. Any isotype
of IgG can be used, including IgG
1, IgG
2, IgG
3, and IgG
4. Variants of the IgG isotypes are also contemplated. The humanized antibody may comprise
sequences from more than one class or isotype. Optimization of the necessary constant
domain sequences to generate the desired biologic activity is readily achieved by
screening the antibodies in the biological assays described in the Examples.
[0089] Likewise, either class of light chain can be used in the compositions and methods
herein. Specifically, kappa, lambda, or variants thereof are useful in the present
compositions and methods.
[0090] Any suitable portion of the CDR sequences from the non-human antibody can be used.
The CDR sequences can be mutagenized by substitution, insertion or deletion of at
least one residue such that the CDR sequence is distinct from the human and non-human
antibody sequence employed. It is contemplated that such mutations would be minimal.
Typically, at least 75% of the humanized antibody residues will correspond to those
of the non-human CDR residues, more often 90%, and most preferably greater than 95%.
[0091] Any suitable portion of the FR sequences from the human antibody can be used. The
FR sequences can be mutagenized by substitution, insertion or deletion of at least
one residue such that the FR sequence is distinct from the human and non-human antibody
sequence employed. It is contemplated that such mutations would be minimal. Typically,
at least 75% of the humanized antibody residues will correspond to those of the human
FR residues, more often 90%, and most preferably greater than 95%, 98% or 99%.
[0092] CDR and FR residues are determined according to the standard sequence definition
of Kabat.
Kabat et al. (1987) Sequences of Proteins of Immunological Interest, National Institutes
of Health, Bethesda Md. SEQ ID NOs: 1-11 show the heavy chain variable domain sequences of various rodent
anti-human GITR antibodies, and SEQ ID NOs: 12-22 depict the light chain variable
domain sequences.
Table 2
| Heavy Chain Sequences and Domains |
| ANTIBODY CLONE |
SEQ ID NO: |
VH RESIDUES |
HEAVY CHAIN CDR RESIDUES |
| CDR-H1 |
CDR-H2 |
CDR-H3 |
| 36H5 |
1 |
1-118 |
26-35 |
50-65 |
98-107 |
| 3D6 |
2 |
1-123 |
26-35 |
50-66 |
99-112 |
| 61G6 |
3 |
1-118 |
26-36 |
51-66 |
99-107 |
| 6H6 |
4 |
1-118 |
26-35 |
50-66 |
99-107 |
| 61F6 |
5 |
1-119 |
26-35 |
50-66 |
99-108 |
| 1D8 |
6 |
1-122 |
26-37 |
52-67 |
100-111 |
| 17F10 |
7 |
1-117 |
26-35 |
50-65 |
98-106 |
| 35D8 |
8 |
1-120 |
26-35 |
50-65 |
98-109 |
| 49A1 |
9 |
1-120 |
26-35 |
50-65 |
98-109 |
| 9E5 |
10 |
1-121 |
26-37 |
52-67 |
100-110 |
| 31H6 |
11 |
1-121 |
26-37 |
52-67 |
100-110 |
Table 3
| Light Chain sequences and Domains |
| ANTIBODY CLONE |
SEQ ID NO: |
VL RESIDUES |
LIGHT CHAIN CDR RESIDUES |
| CDR-L1 |
CDR-L2 |
CDR-L3 |
| 36H5 |
12 |
1-113 |
24-39 |
54-60 |
93-101 |
| 3D6 |
13 |
1-113 |
24-39 |
55-61 |
94-102 |
| 61G6 |
14 |
1-108 |
24-33 |
49-55 |
88-96 |
| 6H6 |
15 |
1-110 |
24-35 |
51-57 |
90-98 |
| 61F6 |
16 |
1-113 |
24-38 |
54-60 |
93-101 |
| 1D8 |
17 |
1-118 |
24-39 |
55-61 |
94-102 |
| 17F10 |
18 |
1-113 |
24-34 |
50-56 |
89-97 |
| 35D8 |
19 |
1-114 |
24-34 |
50-56 |
89-98 |
| 49A1 |
20 |
1-114 |
24-34 |
50-56 |
89-98 |
| 9E5 |
21 |
1-113 |
24-34 |
50-56 |
89-97 |
| 31H6 |
22 |
1-113 |
24-34 |
50-56 |
89-97 |
[0093] In one embodiment, CDRs include variants of any single sequence CDR disclosed herein
(SEQ ID NOs: 23-88), in which the variant comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10
or more conservative amino acid substitutions relative to the disclosed sequence,
as determined using the data of Table 1.
[0094] Also contemplated are chimeric antibodies. As noted above, typical chimeric antibodies
comprise a portion of the heavy and/or light chain identical with, or homologous to,
corresponding sequences in antibodies derived from a particular species or belonging
to a particular antibody class or subclass, while the remainder of the chain(s) is
identical with or homologous to corresponding sequences in antibodies derived from
another species or belonging to another antibody class or subclass, as well as fragments
of such antibodies, so long as they exhibit the desired biological activity.
See U.S. Pat. No. 4,816,567; and
Morrison et al. (1984) Proc. Natl. Acad. Sci. USA 81: 6851-6855.
[0095] Bispecific antibodies are also useful in the present methods and compositions. As
used herein, the term "bispecific antibody" refers to an antibody, typically a monoclonal
antibody, having binding specificities for at least two different antigenic epitopes.
In one embodiment, the epitopes are from the same antigen. In another embodiment,
the epitopes are from two different antigens. Methods for making bispecific antibodies
are known in the art. For example, bispecific antibodies can be produced recombinantly
using the co-expression of two immunoglobulin heavy chain/light chain pairs.
See, e.g., Milstein et al. (1983) Nature 305: 537-39. Alternatively, bispecific antibodies can be prepared using chemical linkage.
See, e.g., Brennan et al. (1985) Science 229:81. Bispecific antibodies include bispecific antibody fragments.
See, e.g., Holliger et al. (1993) Proc. Natl. Acad. Sci. U.S.A. 90:6444-48,
Gruber et al. (1994) J. Immunol. 152:5368.
[0096] In yet other embodiments, different constant domains may be appended to humanized
V
L and V
H regions derived from the CDRs provided herein. For example, if a particular intended
use of an antibody (or fragment) of the present invention were to call for altered
effector functions, a heavy chain constant domain other than IgG1 may be used. Although
IgG1 antibodies provide for long half-life and for effector functions, such as complement
activation and antibody-dependent cellular cytotoxicity, such activities may not be
desirable for all uses of the antibody. In such instances an IgG4 or IgG2 constant
domain, for example, may be used.
[0097] The parental and engineered forms of the antibodies of the present invention may
also be conjugated to a chemical moiety. The chemical moiety may be,
inter alia, a polymer, a radionuclide or a cytotoxic factor. Preferably the chemical moiety
is a polymer which increases the half-life of the antibody molecule in the body of
a subject. Suitable polymers include, but are not limited to, polyethylene glycol
(PEG) (
e.g., PEG with a molecular weight of 2kDa, 5 kDa, 10 kDa, 12kDa, 20 kDa, 30kDa or 40kDa),
dextran and monomethoxypolyethylene glycol (mPEG).
Lee et al., (1999) (Bioconj. Chem. 10:973-981) discloses PEG conjugated single-chain antibodies.
Wen et al., (2001) (Bioconj. Chem. 12:545-553) disclose conjugating antibodies with PEG which is attached to a radiometal chelator
(diethylenetriaminpentaacetic acid (DTPA)).
[0098] The antibodies and antibody fragments or the GITR soluble proteins or fragments thereof
of the invention may also be conjugated with labels such as
99Tc,
90Y,
111In,
32P,
14C,
125I,
3H,
131I,
11C,
15O,
13N,
18F,
35S,
51Cr,
57To,
226Ra,
60Co,
59Fe,
57Se,
152Eu,
67CU,
217Ci,
211At,
212Pb,
47Sc,
109Pd,
234Th, and
40K,
157Gd,
55Mn,
52Tr and
56Fe.
[0099] The antibodies and antibody fragments or the GITR soluble proteins or fragments thereof
of the invention may also be conjugated with fluorescent or chemilluminescent labels,
including fluorophores such as rare earth chelates, fluorescein and its derivatives,
rhodamine and its derivatives, isothiocyanate, phycoerythrin, phycocyanin, allophycocyanin,
o-phthaladehyde, fluorescamine,
152Eu, dansyl, umbelliferone, luciferin, luminal label, isoluminal label, an aromatic
acridinium ester label, an imidazole label, an acridimium salt label, an oxalate ester
label, an aequorin label, 2,3-dihydrophthalazinediones, biotin/avidin, spin labels
and stable free radicals.
V. Biological Activity of Humanized Anti-GITR Antibodies
[0101] Antibodies having the characteristics identified herein as being desirable in a humanized
anti-GITR antibody can be screened for inhibitory biologic activity
in vitro or suitable binding affinity. Agonist antibodies may be distinguished from antagonist
antibodies using the biological assay provided at Example 5. Antibodies that exhibit
agonist activity will not block the activity of GITR, but will instead stimulate the
response typically mediated by GITR signaling.
[0102] To screen for antibodies that bind to the epitope on human GITR bound by an antibody
of interest (e.g., those that block binding of GITR), a routine cross-blocking assay
such as that described in
ANTIBODIES, A LABORATORY MANUAL, Cold Spring Harbor Laboratory, Ed Harlow and David
Lane (1988), can be performed. Antibodies that bind to the same epitope are likely to cross-block
in such assays, but not all cross-blocking antibodies will necessarily bind at precisely
the same epitope since cross-blocking may result from steric hindrance of antibody
binding by antibodies bind at overlapping epitopes, or even nearby non-overlapping
epitopes.
[0103] Alternatively, epitope mapping,
e.g., as described in
Champe et al. (1995) J. Biol. Chem. 270:1388-1394, can be performed to determine whether the antibody binds an epitope of interest.
"Alanine scanning mutagenesis," as described by
Cunningham and Wells (1989) Science 244: 1081-1085, or some other form of point mutagenesis of amino acid residues in human GITR may
also be used to determine the functional epitope for an anti-GITR antibody of the
present invention. Mutagenesis studies, however, may also reveal amino acid residues
that are crucial to the overall three-dimensional structure of GITR but that are not
directly involved in antibody-antigen contacts, and thus other methods may be necessary
to confirm a functional epitope determined using this method.
[0104] The epitope bound by a specific antibody may also be determined by assessing binding
of the antibody to peptides comprising fragments of human GITR (SEQ ID NO: 41). A
series of overlapping peptides encompassing the sequence of GITR may be synthesized
and screened for binding, e.g. in a direct ELISA, a competitive ELISA (where the peptide
is assessed for its ability to prevent binding of an antibody to GITR bound to a well
of a microtiter plate), or on a chip. Such peptide screening methods may not be capable
of detecting some discontinuous functional epitopes, i.e. functional epitopes that
involve amino acid residues that are not contiguous along the primary sequence of
the GITR polypeptide chain.
[0106] With regard to X-ray crystallography, crystallization may be accomplished using any
of the known methods in the art (
e.g. Giege et al. (1994) Acta Crystallogr. D50:339-350;
McPherson (1990) Eur. J. Biochem. 189:1-23), including microbatch (
e.g. Chayen (1997) Structure 5:1269-1274), hanging-drop vapor diffusion (
e.g. McPherson (1976) J. Biol. Chem. 251:6300-6303), seeding and dialysis. It is desirable to use a protein preparation having a concentration
of at least about 1 mg/mL and preferably about 10 mg/mL to about 20 mg/mL. Crystallization
may be best achieved in a precipitant solution containing polyethylene glycol 1000-20,000
(PEG; average molecular weight ranging from about 1000 to about 20,000 Da), preferably
about 5000 to about 7000 Da, more preferably about 6000 Da, with concentrations ranging
from about 10% to about 30% (w/v). It may also be desirable to include a protein stabilizing
agent, e.g. glycerol at a concentration ranging from about 0.5% to about 20%. A suitable
salt, such as sodium chloride, lithium chloride or sodium citrate may also be desirable
in the precipitant solution, preferably in a concentration ranging from about 1 mM
to about 1000 mM. The precipitant is preferably buffered to a pH of from about 3.0
to about 5.0, preferably about 4.0. Specific buffers useful in the precipitant solution
may vary and are well-known in the art. Scopes, Protein Purification: Principles and
Practice, Third ed., (1994) Springer-Verlag, New York. Examples of useful buffers
include, but are not limited to, HEPES, Tris, MES and acetate. Crystals may be grow
at a wide range of temperatures, including 2°C, 4°C, 8°C and 26°C.
[0108] Additional antibodies binding to the same epitope as an antibody of the present invention
may be obtained, for example, by screening of antibodies raised against GITR for binding
to the epitope, or by immunization of an animal with a peptide comprising a fragment
of human GITR comprising the epitope sequence. Antibodies that bind to the same functional
epitope might be expected to exhibit similar biological activities, such as blocking
receptor binding, and such activities can be confirmed by functional assays of the
antibodies.
[0109] Antibody affinities may be determined using standard analysis. Preferred humanized
antibodies are those that bind human GITR with a K
d value of no more than about 1x10
-7; preferably no more than about 1x10
-8; more preferably no more than about 1x10
-9; and most preferably no more than about 1x10
-10 or even 1x10
-11 M.
[0110] The antibodies and fragments thereof useful in the present compositions and methods
are biologically active antibodies and fragments. As used herein, the term "biologically
active" refers to an antibody or antibody fragment that is capable of binding the
desired the antigenic epitope and directly or indirectly exerting a biologic effect.
As used herein, the term "specific" refers to the selective binding of the antibody
to the target antigen epitope. Antibodies can be tested for specificity of binding
by comparing binding to GITR to binding to irrelevant antigen or antigen mixture under
a given set of conditions. If the antibody binds to GITR at least 10, and preferably
50 times more than to irrelevant antigen or antigen mixture then it is considered
to be specific. An antibody that "specifically binds" to GITR does not bind to proteins
that do not comprise the GITR-derived sequences, i.e. "specificity" as used herein
relates to GITR specificity, and not any other sequences that may be present in the
protein in question. For example, as used herein, an antibody that "specifically binds"
to a polypeptide comprising GITR will typically bind to FLAG
®-GITR, which is a fusion protein comprising GITR and a FLAG
® peptide tag, but it does not bind to the FLAG
® peptide tag alone or when it is fused to a protein other than GITR.
[0111] GITR-specific binding compounds of the present invention, such as agonistic GITR
specific antibodies, can enhance its biological activity in any manner, including
but not limited to increasing the immune response to a microbial infection.
VI. Pharmaceutical Compositions
[0113] Formulations of therapeutic and diagnostic agents may be prepared by mixing with
physiologically acceptable carriers, excipients, or stabilizers in the form of, e.g.,
lyophilized powders, slurries, aqueous solutions or suspensions.
See, e.g., Hardman et al. (2001) Goodman and Gilman's The Pharmacological Basis of Therapeutics,
McGraw-Hill, New York, NY;
Gennaro (2000) Remington: The Science and Practice of Pharmacy, Lippincott, Williams,
and Wilkins, New York, NY;
Avis et al. (eds.) (1993) Pharmaceutical Dosage Forms: Parenteral Medications, Marcel
Dekker, NY;
Lieberman, et al. (eds.) (1990) Pharmaceutical Dosage Forms: Tablets, Marcel Dekker,
NY;
Lieberman et al. (eds.) (1990) Pharmaceutical Dosage Forms: Disperse Systems, Marcel
Dekker, NY;
Weiner and Kotkoskie (2000) Excipient Toxicity and Safety, Marcel Dekker, Inc., New
York, NY.
[0114] Toxicity and therapeutic efficacy of the antibody compositions, administered alone
or in combination with an immunosuppressive agent, can be determined by standard pharmaceutical
procedures in cell cultures or experimental animals, e.g., for determining the LD
50 (the dose lethal to 50% of the population) and the ED
50 (the dose therapeutically effective in 50% of the population). The dose ratio between
toxic and therapeutic effects is the therapeutic index and it can be expressed as
the ratio of LD
50 to ED
50. Antibodies exhibiting high therapeutic indices are preferred. The data obtained
from these cell culture assays and animal studies can be used in formulating a range
of dosage for use in human. The dosage of such compounds lies preferably within a
range of circulating concentrations that include the ED
50 with little or no toxicity. The dosage may vary within this range depending upon
the dosage form employed and the route of administration.
[0115] The mode of administration is not particularly important. Suitable routes of administration
may, for example, include oral, rectal, transmucosal, or intestinal administration;
parenteral delivery, including intramuscular, subcutaneous, intramedullary injections,
as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal,
or intraocular injections. Administration of antibody used in the pharmaceutical composition
or to practice the method of the present invention can be carried out in a variety
of conventional ways, such as oral ingestion, inhalation, topical application or cutaneous,
subcutaneous, intraperitoneal, parenteral, intraarterial or intravenous injection.
[0116] Alternately, one may administer the antibody in a local rather than systemic manner,
for example, via injection of the antibody directly into an arthritic joint or pathogen-induced
lesion characterized by immunopathology, often in a depot or sustained release formulation.
Furthermore, one may administer the antibody in a targeted drug delivery system, for
example, in a liposome coated with a tissue-specific antibody, targeting, for example,
arthritic joint or pathogen-induced lesion characterized by immunopathology. The liposomes
will be targeted to and taken up selectively by the afflicted tissue.
[0117] Selecting an administration regimen for a therapeutic depends on several factors,
including the serum or tissue turnover rate of the entity, the level of symptoms,
the immunogenicity of the entity, and the accessibility of the target cells in the
biological matrix. Preferably, an administration regimen maximizes the amount of therapeutic
delivered to the patient consistent with an acceptable level of side effects. Accordingly,
the amount of biologic delivered depends in part on the particular entity and the
severity of the condition being treated. Guidance in selecting appropriate doses of
antibodies, cytokines, and small molecules are available.
See, e.g., Wawrzynczak (1996) Antibody Therapy, Bios Scientific Pub. Ltd, Oxfordshire, UK;
Kresina (ed.) (1991) Monoclonal Antibodies, Cytokines and Arthritis, Marcel Dekker,
New York, NY;
Bach (ed.) (1993) Monoclonal Antibodies and Peptide Therapy in Autoimmune Diseases,
Marcel Dekker, New York, NY;
Baert et al. (2003) New Engl. J. Med. 348:601-608;
Milgrom et al. (1999) New Engl. J. Med. 341:1966-1973;
Slamon et al. (2001) New Engl. J. Med. 344:783-792;
Beniaminovitz et al. (2000) New Engl. J. Med. 342:613-619;
Ghosh et al. (2003) New Engl. J. Med. 348:24-32;
Lipsky et al. (2000) New Engl. J. Med. 343:1594-1602.
[0118] Determination of the appropriate dose is made by the clinician, e.g., using parameters
or factors known or suspected in the art to affect treatment or predicted to affect
treatment. Generally, the dose begins with an amount somewhat less than the optimum
dose and it is increased by small increments thereafter until the desired or optimum
effect is achieved relative to any negative side effects. Important diagnostic measures
include those of symptoms of, e.g., the inflammation or level of inflammatory cytokines
produced. Preferably, a biologic that will be used is substantially derived from the
same species as the animal targeted for treatment (e.g. a humanized antibody for treatment
of human subjects), thereby minimizing any immune response to the reagent.
[0119] Antibodies, antibody fragments, and cytokines can be provided by continuous infusion,
or by doses at intervals of, e.g., one day, 1-7 times per week, one week, two weeks,
monthly, bimonthly, etc. Doses may be provided intravenously, subcutaneously, topically,
orally, nasally, rectally, intramuscular, intracerebrally, intraspinally, or by inhalation.
A preferred dose protocol is one involving the maximal dose or dose frequency that
avoids significant undesirable side effects. A total weekly dose is generally at least
0.05 µg/kg, 0.2 µg/kg, 0.5 µg/kg, 1 µg/kg, 10 µg/kg, 100 µg/kg, 0.2 mg/kg, 1.0 mg/kg,
2.0 mg/kg, 10 mg/kg, 25 mg/kg, 50 mg/kg body weight or more.
See, e.g., Yang et al. (2003) New Engl. J. Med. 349:427-434;
Herold et al. (2002) New Engl. J. Med. 346:1692-1698;
Liu et al. (1999) J. Neurol. Neurosurg. Psych. 67:451-456;
Portielji et al. (20003) Cancer Immunol. Immunother. 52:133-144. The desired dose of a small molecule therapeutic, e.g., a peptide mimetic, natural
product, or organic chemical, is about the same as for an antibody or polypeptide,
on a moles/kg basis.
[0120] As used herein, "inhibit" or "treat" or "treatment" includes a postponement of development
of the symptoms associated with autoimmune disease or pathogen-induced immunopathology
and/or a reduction in the severity of such symptoms that will or are expected to develop.
The terms further include ameliorating existing uncontrolled or unwanted autoimmune-related
or pathogen-induced immunopathology symptoms, preventing additional symptoms, and
ameliorating or preventing the underlying causes of such symptoms. Thus, the terms
denote that a beneficial result has been conferred on a vertebrate subject with an
autoimmune or pathogen-induced immunopathology disease or symptom, or with the potential
to develop such a disease or symptom.
[0121] As used herein, the term "therapeutically effective amount" or "effective amount"
refers to an amount of an GITR-specific binding compound, e.g. and antibody, that
when administered alone or in combination with an additional therapeutic agent to
a cell, tissue, or subject is effective to prevent or ameliorate the autoimmune disease
or pathogen-induced immunopathology associated disease or condition or the progression
of the disease. A therapeutically effective dose further refers to that amount of
the compound sufficient to result in amelioration of symptoms, e.g., treatment, healing,
prevention or amelioration of the relevant medical condition, or an increase in rate
of treatment, healing, prevention or amelioration of such conditions. When applied
to an individual active ingredient administered alone, a therapeutically effective
dose refers to that ingredient alone. When applied to a combination, a therapeutically
effective dose refers to combined amounts of the active ingredients that result in
the therapeutic effect, whether administered in combination, serially or simultaneously.
An effective amount of therapeutic will decrease the symptoms typically by at least
10%; usually by at least 20%; preferably at least about 30%; more preferably at least
40%, and most preferably by at least 50%.
[0122] Methods for co-administration or treatment with a second therapeutic agent, e.g.,
a cytokine, antibody, steroid, chemotherapeutic agent, antibiotic, anti-viral, or
radiation, are well known in the art, see, e.g.,
Hardman et al. (eds.) (2001) Goodman and Gilman's The Pharmacological Basis of Therapeutics,
10th ed., McGraw-Hill, New York, NY;
Poole and Peterson (eds.) (2001) Pharmacotherapeutics for Advanced Practice: A Practical
Approach, Lippincott, Williams & Wilkins, Phila., PA;
Chabner and Longo (eds.) (2001) Cancer Chemotherapy and Biotherapy, Lippincott, Williams
& Wilkins, Phila., PA.
[0123] Chemotherapeutic agents include alkylating agents such as thiotepa and CYTOXAN® cyclosphosphamide;
alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as
benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines
including altretamine, triethylenemelamine, trietylenephosphoramide, triethiylenethiophosphoramide
and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a
camptothecin (including the synthetic analogue topotecan); bryostatin; callystatin;
CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues);
cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin
(including the synthetic analogues, KW-2189 and CB 1-TM1); eleutherobin; pancratistatin;
a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine,
cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide
hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide,
uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine,
nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e.g., calicheamicin,
especially calicheamicin gammall and calicheamicin omegall (see, e.g.,
Agnew, Chem. Intl. Ed. Engl., 33: 183-186 (1994)); dynemicin, including dynemicin A; bisphosphonates, such as clodronate; an esperamicin;
as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic
chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin,
carabicin, caminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin,
detorubicin, 6-diazo-5-oxo-L-norleucine, ADRIAMYCIN® doxorubicin (including morpholino-doxorubicin,
cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin,
esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic
acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin,
streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites
such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin,
methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine,
thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine,
carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens
such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone;
anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher
such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid;
eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine;
diaziquone; elformithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate;
hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins;
mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin;
losoxantrone; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK® polysaccharide
complex (JHS Natural Products, Eugene, Oreg.); razoxane; rhizoxin; sizofuran; spirogermanium;
tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylamine; trichothecenes (especially
T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine; dacarbazine;
mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C");
cyclophosphamide; thiotepa; taxoids, e.g., TAXOL® paclitaxel (Bristol-Myers Squibb
Oncology, Princeton, N.J.), ABRAXANE™ Cremophor-free, albumin-engineered nanoparticle
formulation of paclitaxel (American Pharmaceutical Partners, Schaumberg, Ill.), and
TAXOTERE® doxetaxel (Rhône-Poulenc Rorer, Antony, France); chloranbucil; GEMZAR® gemcitabine;
6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and
carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitoxantrone; vincristine;
NAVELBINE® vinorelbine; novantrone; teniposide; edatrexate; daunomycin; aminopterin;
XELODA® capecitabine; ibandronate; CPT-11; topoisomerase inhibitor RFS 2000; difluoromethylornithine
(DMFO); retinoids such as retinoic acid; and pharmaceutically acceptable salts, acids
or derivatives of any of the above.
[0124] Also included are anti-hormonal agents that act to regulate or inhibit hormone action
on tumors such as anti-estrogens and selective estrogen receptor modulators (SERMs),
including, for example, tamoxifen (including NOLVADEX® tamoxifen), raloxifene, droloxifene,
4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and FARESTON. toremifene;
aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production
in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE®
megestrol acetate, AROMASIN® exemestane, formestanie, fadrozole, RIVISOR® vorozole,
FEMARA® letrozole, and ARIMIDEX® anastrozole; and anti-androgens such as flutamide,
nilutamide, bicalutamide, leuprolide, and goserelin; as well as troxacitabine (a 1,3-dioxolane
nucleoside cytosine analog); antisense oligonucleotides, particularly those which
inhibit expression of genes in signaling pathways implicated in abherant cell proliferation,
such as, for example, PKC-alpha, Ralf and H-Ras; ribozymes such as a VEGF expression
inhibitor (e.g., ANGIOZYME® ribozyme) and a HER2 expression inhibitor; vaccines such
as gene therapy vaccines, for example, ALLOVECTIN® vaccine, LEUVECTIN® vaccine, and
VAXID® vaccine; PROLEUKIN® rIL-2; LURTOTECAN® topoisomerase 1 inhibitor; ABARELIX®
rmRH; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
[0125] In particular, transforming growth factor (TGF)-β displays an array of pleiotropic
effects in cellular functions such as proliferation, homeostasis, angiogenesis and
wound healing. Aberrant regulation of TGF-β function contributes to cancer progression.
Most cancers are characterized by excessive transforming growth factor-β production
by tumors, which can promote tumor growth and mediate epithelial-to-mesenchymal transition.
TGF-β also plays a pivotal role within the immune system maintaining tolerance via
the regulation of lymphocyte proliferation, differentiation, and survival. TGF-β has
been proven to be an important suppressive element in enhancing Treg function and
dampening tumor immunity. Administration of TGF-β inhibitors in conjunction with GITR
agonists, e.g., antibodies, is contemplated.
[0126] Also contemplated is co-administration with anti-viral therapeutics. Anti-virals
include any drug that destroys viruses. Antivirals may include interferons which function
to inhibits replication of the virus, protease inhibitors, and reverse transcriptase
inhibitors or agents contained in the combination of highly active antiretroviral
therapy (HAART) for HIV.
[0127] Typical veterinary, experimental, or research subjects include monkeys, dogs, cats,
rats, mice, rabbits, guinea pigs, horses, and humans.
VII. Antibody Production
[0128] In one embodiment, for recombinant production of the antibodies of the present invention,
the nucleic acids encoding the two chains are isolated and inserted into one or more
replicable vectors for further cloning (amplification of the DNA) or for expression.
DNA encoding the monoclonal antibody is readily isolated and sequenced using conventional
procedures (e.g., by using oligonucleotide probes that are capable of binding specifically
to genes encoding the heavy and light chains of the antibody). Many vectors are available.
The vector components generally include, but are not limited to, one or more of the
following: a signal sequence, an origin of replication, one or more marker genes,
an enhancer element, a promoter, and a transcription termination sequence. In one
embodiment, both the light and heavy chains of a humanized anti-GITR antibody of the
present invention are expressed from the same vector, e.g. a plasmid or an adenoviral
vector.
[0129] Antibodies of the present invention may be produced by any method known in the art.
In one embodiment, antibodies are expressed in mammalian or insect cells in culture,
such as Chinese hamster ovary (CHO) cells, human embryonic kidney (HEK) 293 cells,
mouse myeloma NSO cells, baby hamster kidney (BHK) cells,
Spodoptera frugiperda ovarian (Sf9) cells. In one embodiment, antibodies secreted from CHO cells are recovered
and purified by standard chromatographic methods, such as protein A, cation exchange,
anion exchange, hydrophobic interaction, and hydroxyapatite chromatography. Resulting
antibodies are concentrated and stored in 20 mM sodium acetate, pH 5.5.
[0130] In another embodiment, the antibodies of the present invention are produced in yeast
according to the methods described in
WO2005/040395. Briefly, vectors encoding the individual light or heavy chains of an antibody of
interest are introduced into different yeast haploid cells, e.g. different mating
types of the yeast
Pichia pastoris, which yeast haploid cells are optionally complementary auxotrophs. The transformed
haploid yeast cells can then be mated or fused to give a diploid yeast cell capable
of producing both the heavy and the light chains. The diploid strain is then able
to secret the fully assembled and biologically active antibody. The relative expression
levels of the two chains can be optimized, for example, by using vectors with different
copy number, using transcriptional promoters of different strengths, or inducing expression
from inducible promoters driving transcription of the genes encoding one or both chains.
[0131] In one embodiment, the respective heavy and light chains of a plurality of different
anti-GITR antibodies (the "original" antibodies) are introduced into yeast haploid
cells to create a library of haploid yeast strains of one mating type expressing a
plurality of light chains, and a library of haploid yeast strains of a different mating
type expressing a plurality of heavy chains. These libraries of haploid strains can
be mated (or fused as spheroplasts) to produce a series of diploid yeast cells expressing
a combinatorial library of antibodies comprised of the various possible permutations
of light and heavy chains. The combinatorial library of antibodies can then be screened
to determine whether any of the antibodies has properties that are superior (e.g.
higher affinity for GITR) to those of the original antibodies.
See. E.g., WO2005/040395.
[0132] In another embodiment, antibodies of the present invention are human domain antibodies
in which portions of an antibody variable domain are linked in a polypeptide of molecular
weight approximately 13 kDa. See, e.g.,
U.S. Pat. Publication No. 2004/0110941. Such single domain, low molecular weight agents provide numerous advantages in terms
of ease of synthesis, stability, and route of administration.
VIII. Uses
[0133] The present invention provides methods for using anti-GITR antibodies and fragments
thereof for the treatment and diagnosis of proliferative or inflammatory disorders
and conditions.
[0134] The present invention provides methods for diagnosing the presence of a microbial
infection or cancer by analyzing expression levels of GITR in test cells, tissue or
bodily fluids compared with GITR levels in cells, tissues or bodily fluids of preferably
the same type from a control. As demonstrated herein, an increase in level of GITR
expression, for example, in the patient versus the control is associated with the
presence of cancer.
[0135] Typically, for a quantitative diagnostic assay, a positive result indicating the
patient tested has cancer or an infectious disease, is one in which the cells, tissues,
or bodily fluids has an GITR expression level at least two times higher, five times
higher, ten times higher, fifteen times higher, twenty times higher, twenty-five times
higher.
[0136] Assay techniques that may be used to determine levels of gene and protein expression,
such as GITR, of the present inventions, in a sample derived from a host are well
known to those of skill in the art. Such assay methods include radioimmunoassays,
reverse transcriptase PCR (RT-PCR) assays, quantitative real-time PCR assays, immunohistochemistry
assays,
in situ hybridization assays, competitive-binding assays, western blot assays, ELISA assays,
and flow cytometric assays, for example, two color FACS analysis for M2 versus M1
phenotyping of tumor-associated macrophages (
Mantovani et al., (2002) TRENDS in Immunology 23:549-555).
[0137] An ELISA assay initially comprises preparing an antibodies of the present invention,
specific to GITR, preferably 36E5, 3D6, 61G6, 6H6, 61F6,1D8, 17F10, 35D8, 49A1, 9E5,
and 31H6 (collectively "GITR antibodies"). In addition, a reporter antibody generally
is prepared that binds specifically to GITR. The reporter antibody is attached to
a detectable reagent such as radioactive, fluorescent or an enzymatic reagent, for
example horseradish peroxidase enzyme or alkaline phosphatase.
[0138] To carry out the ELISA, at least one of the GITR antibodies described above is incubated
on a solid support, e.g., a polystyrene dish that binds the antibody. Any free protein
binding sites on the dish are then covered by incubating with a non-specific protein,
such as bovine serum albumin. Next, the sample to be analyzed is incubated in the
dish, during which time GITR binds to the specific GITR antibody attached to the polystyrene
dish. Unbound sample is washed out with buffer. A reporter antibody specifically directed
to GITR and linked to horseradish peroxidase is placed in the dish resulting in binding
of the reporter antibody to any monoclonal antibody bound to GITR. Unattached reporter
antibody is then washed out. Reagents for peroxidase activity, including a calorimetric
substrate are then added to the dish. Immobilized peroxidase, linked to GITR antibodies,
produces a colored reaction product. The amount of color developed in a given time
period is proportional to the amount of GITR protein present in the sample. Quantitative
results typically are obtained by reference to a standard curve.
[0139] A competition assay may be employed wherein antibodies specific to GITR are attached
to a solid support and labeled GITR and a sample derived from the host are passed
over the solid support and the amount of label detected attached to the solid support
can be correlated to a quantity of GITR in the sample.
[0140] The above tests may be carried out on samples derived from a variety of cells, bodily
fluids and/or tissue extracts such as homogenates or solubilized tissue obtained from
a patient. Tissue extracts are obtained routinely from tissue biopsy and autopsy material.
Bodily fluids useful in the present invention include blood, urine, saliva or any
other bodily secretion or derivative thereof. The term "blood" is meant to include
whole blood, plasma, serum or any derivative of blood.
[0141] Antibodies of the present invention may be used to treat viral infections. HIV infection
is characterized by defects in the generation and maintenance of central memory cells.
CD8+ central memory cells have a shorter half-life and are less abundant in HIV-infected
individuals than in controls. Also, the frequency of both CD4+ and CD8+ HIV-specific
T cells decreases rapidly after initiation of highly active antiretroviral therapy
(HAART). Co-stimulation on CD4+ by anti-GITR may provide a mechanism to increase memory
CD8+ response and to contribute to clearance of the virus. It has been shown that
treatment of persistently Friend virus-infected mice with anti-GITR antibody to ameliorate
suppression by Tregs significantly improved IFN-γ production by the CD8+ T cells and
allowed a significant reduction in viral loads (
Dittmer et al., (2004) Immunity 20: 293-303).
[0142] Another characteristic of HIV infection is massive apoptosis of CD4+ T cells starting
early in HIV infection. The progressive apoptotic deletion of CD4 T cells contributes
to weakened HIV-specific cellular immune responses and to the development of AIDS.
GITR co-stimulation has been shown to enhance murine antigen-specific cytokine secretion
by protecting T cells from apoptosis.
Lahey et al. (2007) J Infect Dis. 196: 43-49) demonstrated that anti-GITR treatment of HIV-specific CD4+ T cells enhances their
cytokine expression and protects them from apoptosis.
[0143] For infections resulting from viral causes, the antibodies of the invention may be
combined by application simulatenous with, prior to or subsequent to application of
standard therapies for treating viral infections. Such standard therapies vary depending
upon type of virus, although in almost all cases, administration of human serum containing
antitibodies (e.g., IgA, IgG) specific to the virus can be effective.
[0144] Influenza infection results in fever, cough, myalgia, headache and malaise, which
often occur in seasonal epidemics. Influenza is also associated with a number of postinfectious
disorders, such as encephalitis, myopericarditis, Goodpasture's syndrome, and Reye's
syndrome. Influenza infection also suppresses normal pulmonary antibacterial defenses,
such that patient's recovering from influenza have an increased risk of developing
bacterial pneumonia.
[0145] Influenza viral surface proteins show marked antigenic variation, resulting from
mutation and recombination. Thus, cytolytic T lymphocytes are the host's primary vehicle
for the elimination of virus after infection. Influenza is classified into three primary
types: A, B and C. Influenza A is unique in that it infects both humans and many other
animals (e.g., pigs, horses, birds and seals) and is the principal cause of pandemic
influenza. Also, when a cell is infected by two different influenza A strains, the
segmented RNA genomes of two two parental virus types mix during replication to create
a hybrid replicant, resulting in new epidemic strains. Influenza B does not replicate
in animals and thus has less genetic variation and influenza C has only a single serotype.
[0146] Most conventional therapies are palliatives of the symptoms resulting from infection,
while the host's immune response actually clears the disease. However, certain strains
(e.g., influenza A) can cause more serious illness and death. Influenza A may be treated
both clinically and prophylactically by the administration of the cyclic amines inhibitors
amantadine and rimantadine, which inhibit viral replication. However, the clinical
utility of these drugs is limited due to the relatively high incidence of adverse
reactions, their narrow anti-viral spectrum (influenza A only), and the propensity
of the virus to become resistant. The administration of serum IgG antibody to the
major influenza surface proteins, hemagglutinin and neuraminidase can prevent pulmonary
infection, whereas mucosal IgA is required to prevent infection of the upper respiratory
tract and trachea. The most effective current treatment for influenza is vaccination
with the administration of virus inactivated with formalin or β-propiolactone.
[0147] After an incubation of 9-11 days, hosts infected with the measles virus develope
fever, cough, coryza and conjunctivitis. Within 1-2 days, an erythematous, maculopapular
rash develop, which quickly spreads over the entire body. Because infection also suppresses
cellular immunity, the host is at greater risk for developing bacterial superinfections,
including otitis media, pneumonia and postinfectious encephalomyelitis. Acute infection
is associated with significant morbidity and mortality, especially in malnourished
adolescents.
[0148] Treatment for measles includes the passive administration of pooled human IgG, which
can prevent infection in non-immune subjects, even if given up to one week after exposure.
[0149] However, prior immunization with live, attenuated virus is the most effective treatment
and prevents disease in more than 95% of those immunized. As there is one serotype
of this virus, a single immunization or infection typically results in protection
for life from subsequent infection.
[0150] In a small proportion of infected hosts, measles can develop into SSPE, which is
a chronic progressive neurologic disorder resulting from a persistent infection of
the central nervous system. SSPE is caused by clonal variants of measles virus with
defects that interfere with virion assembly and budding. For these patients, reactivation
of T-cells with the antibodies of the invention so as to facilitate viral clearance
would be desirable.
[0151] Hepatitis B virus (HB-V) is the most infectious known bloodborne pathogen. It is
a major cause of acute and chronic heptatis and hepatic carcinoma, as well as life-long,
chronic infection. Following infection, the virus replicates in hepatocytes, which
also then shed the surface antigen HBsAg. The detection of excessive levels of HBsAg
in serum is used a standard method for diagnosing a hepatitis B infection. An acute
infection may resolve or it can develop into a chronic persistent infection.
[0152] Current treatments for chronic HBV include α-inteferon, which increases the expression
of class I human leukocyte antigen (HLA) on the surface of hepatocytes, thereby facilitating
their recognition by cytotoxic T lymphocytes. Additionally, the nucleoside analogs
ganciclovir, famciclovir and lamivudine have also shown some efficacy in the treatment
of HBV infection in in clinical trial. Additional treatments for HBV include pegylated
α-interferon, adenfovir, entecavir and telbivudine. While passive immunity can be
conferred through parental administration of anti-HBsAg serum antibodies, vaccination
with inactivated or recombinant HBsAg also confers resistance to infection. The antibodies
of the invention may be combined with conventional treatments for hepatitis B infections
for therapeutic advantage.
[0153] Hepatitis C virus (HC-V) infection may lead to a chronic form of hepatitis, resulting
in cirrosis. While symptoms are similar to infections resulting from Hepatitis B,
in distinct contrast to HB-V, infected hosts can be asymptomatic for 10-20 years.
Treatment for HC-V infection includes the administration of a combination of α-interferon
and ribavirin. A promising potential therapy for HC-V infection is the protease inhibitor
telaprevir (VX-960). Additional treatments include: anti-PD-1 antibody (MDX-1106,
Medarex), bavituximab (an antibody that binds anionic phospholipid phosphatidylserine
in a B2-glycoprotein I dependent manner, Peregrine Pharmaceuticals), anti-HPV viral
coat protein E2 antibod(y)(ies) (E.g., ATL 6865-Ab68+Ab65, XTL Pharmaceuticals) and
Civacir® (polyclonal anti-HCV human immune globulin). The antibodies of the invention
may be combined with one or more of these treatments for hepatitis C infections for
therapeutic advantage.
[0154] The broad scope of this invention is best understood with reference to the following
examples, which are not intended to limit the inventions to the specific embodiments.
The specific embodiments described herein are offered by way of example only, and
the invention is to be limited by the terms of the appended claims, along with the
full scope of equivalents to which such claims are entitled.
EXAMPLES
Example 1
General Methods
[0155] Standard methods in molecular biology are described.
Maniatis et al. (1982) Molecular Cloning, A Laboratory Manual, Cold Spring Harbor
Laboratory Press, Cold Spring Harbor, NY;
Sambrook and Russell (2001) Molecular Cloning, 3rd ed., Cold Spring Harbor Laboratory
Press, Cold Spring Harbor, NY;
Wu (1993) Recombinant DNA, Vol. 217, Academic Press, San Diego, CA. Standard methods also appear in
Ausbel et al. (2001) Current Protocols in Molecular Biology, Vols. 1-4, John Wiley
and Sons, Inc. New York, NY, which describes cloning in bacterial cells and DNA mutagenesis (Vol. 1), cloning
in mammalian cells and yeast (Vol. 2), glycoconjugates and protein expression (Vol.
3), and bioinformatics (Vol. 4).
[0156] Methods for protein purification including immunoprecipitation, chromatography, electrophoresis,
centrifugation, and crystallization are described.
Coligan et al. (2000) Current Protocols in Proteins Science, Vol. 1, John Wiley and
Sons, Inc., New York. Chemical analysis, chemical modification, post-translational modification, production
of fusion proteins, glycosylation of proteins are described.
See, e.g., Coligan et al. (2000) Current Protocols in Protein Science, Vol. 2, John Wiley and
Sons, Inc., New York;
Ausubel et al. (2001) Current Protocols in Molecular Biology, Vol. 3, John Wiley and
Sons, Inc., NY, NY, pp. 16.0.5-16.22.17;
Sigma-Aldrich, Co. (2001) Products for Life Science Research, St. Louis, MO; pp. 45-89;
Amersham Pharmacia Biotech (2001) BioDirectory, Piscataway, N.J., pp. 384-391. Production, purification, and fragmentation of polyclonal and monoclonal antibodies
are described.
Coligan et al. (2001) Current Protcols in Immunology, Vol. 1, John Wiley and Sons,
Inc., New York;
Harlow and Lane (1999) Using Antibodies, Cold Spring Harbor Laboratory Press, Cold
Spring Harbor, NY; Harlow and Lane,
supra. Standard techniques for characterizing ligand/receptor interactions are available.
See, e.g., Coligan et al. (2001) Current Protcols in Immunology, Vol. 4, John Wiley, Inc., New
York.
[0157] Methods for flow cytometry, including fluorescence activated cell sorting detection
systems (FACS®), are available.
See, e.g., Owens et al. (1994) Flow Cytometry Principles for Clinical Laboratory Practice, John
Wiley and Sons, Hoboken, NJ;
Givan (2001) Flow Cytometry, 2nd ed.; Wiley-Liss, Hoboken, NJ;
Shapiro (2003) Practical Flow Cytometry, John Wiley and Sons, Hoboken, NJ. Fluorescent reagents suitable for modifying nucleic acids, including nucleic acid
primers and probes, polypeptides, and antibodies, for use, e.g., as diagnostic reagents,
are available.
Molecular Probes (2003) Catalogue, Molecular Probes, Inc., Eugene, OR ;
Sigma-Aldrich (2003) Catalogue, St. Louis, MO.
[0158] Standard methods of histology of the immune system are described.
See, e.g., Muller-Harmelink (ed.) (1986) Human Thymus: Histopathology and Pathology, Springer
Verlag, New York, NY;
Hiatt, et al. (2000) Color Atlas of Histology, Lippincott, Williams, and Wilkins,
Phila, PA;
Louis, et al. (2002) Basic Histology: Text and Atlas, McGraw-Hill, New York, NY.
[0159] Software packages and databases for determining, e.g., antigenic fragments, leader
sequences, protein folding, functional domains, glycosylation sites, and sequence
alignments, are available.
See, e.g., GenBank, Vector NTI® Suite (Informax, Inc, Bethesda, MD); GCG Wisconsin Package (Accelrys,
Inc., San Diego, CA); DeCypher® (TimeLogic Corp., Crystal Bay, Nevada);
Menne et al. (2000) Bioinformatics 16: 741-742;
Menne et al. (2000) Bioinformatics Applications Note 16:741-742;
Wren et al. (2002) Comput. Methods Programs Biomed. 68:177-181;
von Heijne (1983) Eur. J. Biochem. 133:17-21;
von Heijne (1986) Nucleic Acids Res. 14:4683-4690.
Example 2
Humanization of Anti-human GITR Antibodies
[0163] Once the target amino acid sequences of the variable heavy and light chains are determined,
plasmids encoding the full-length humanized antibody may be generated. Plasmid sequences
may be altered using Kunkel mutagenesis (see, e.g.,
Kunkel T A. (1985) Proc. Natl. Acad. Sci. U.S.A 82:488-492) to change the DNA sequence to the target humanized antibody sequences. Simultaneously,
codon optimization may be performed to provide for potentially optimal expression.
[0164] Antibodies of the present invention can be humanized using a method that identifies
an acceptor germline sequence for a humanized antibody, and comprises the steps of:
a) identifying a non-human antibody that has the desired biological activity; b) determining
the amino acid sequence of a non-human antibody V
H and V
L domains; and c) comparing the nonhuman antibody sequence to a group of human germline
sequences, wherein the comparison comprises the substeps of: 1) assigning the non-human
V sequences residue numbers according to Kabat
supra; 2) delineating the CDR and FR regions in the sequence according to Kabat
supra; 3) assigning a predetermined numerical score at specific residue position for which
the non-human and human antibody germline sequences are identical; and 4) totaling
all of the residue scores to generate a total score for each human germline sequence;
and d) identifying the human germline sequence with the highest total residue score
as the acceptor germline sequence. In one embodiment, the method further comprises
the substeps of: 5) assigning a numerical score of 1 for each FR residue position
for which the non-human and human antibody germline sequences are identical that was
not scored in substep (3) to germline sequences with identical total residue scores
after substep (4); 6) totaling all of the residue scores to generate a total score
for each human germline sequence. In a specific embodiment, the non-human antibody
is specific for GITR and enhances the biological activity of GITR. Also provided herein
is an antibody generated by the above method.
[0165] In one embodiment, the GITR antibody is humanized using the following method. First,
the non-human V
L and V
H domains of the GITR antibody are cloned and sequenced, and the amino acid sequence
determined. Then, the non-human V
H sequence are compared to a group of three human V
H germline amino acid sequences. The three groups contain one representative from each
of subgroups IGHV1, IGHV3 and IGHV4. The V
H subgroups are listed in
M.-P. Lefranc, Exp. Clin. Immunogenetics, 18:100-116 (2001). Specifically, the comparison with the three germline sequences begins with the
assignment of residue numbers to the non-human V
H sequence according to the Kabat numbering system.
See Kabat, et al., U. S. Department of Health and Human Services, NIH Pub. 91-3242 (5th
Ed., 1991). The non-human V
H sequence are then aligned with each of the three human germline sequences. Since
the V genes only comprise V
H residues 1-94, only these residues are considered in the alignment. Next, the complementarity-determining
(CDR) and framework (FR) regions in the sequence are delineated. CDR and FR are delineated
according to the combination of the definitions provided in Kabat, et al., U. S. Department
of Health and Human Services, NIH Pub. 91-3242 (5th Ed., 1991), and
C. Chothia & A.M. Lesk, J. Mol. Biol., 196:901-917 (1987). Therefore, the CDR definition used is residues 26-35 for CDR1, residues 50-65 for
CDR2, and CDR3 is residues 95-102 for CDR3 of the V
H domain. The next step involves assigning a numerical score at identified residue
position where the non-human and human sequences are identical. One example of this
scoring is shown in Table 4 below.
| Table 4 |
| Residue # |
Score |
Reason |
| 24 |
3 |
Affects CDR-H1 |
| 27 |
4 |
Affects CDR-H1,3* |
| 29 |
4 |
Affects CDR-H1* |
| 34 |
4 |
Affects CDR-H1* |
| 35 |
2 |
VH/VL interface |
| 37 |
2 |
VH/VL interface |
| 48 |
3 |
Affects CDR-H2 |
| 49 |
3 |
Affects CDR-H2 |
| 50 |
2 |
VH/VL interface |
| 58 |
2 |
VH/VL interface |
| 60 |
2 |
VH/VL interface |
| 63 |
3 |
Affects CDR-H2 |
| 67 |
3 |
Affects CDR-H2 |
| 69 |
3 |
Affects CDR-H2 |
| 71 |
4 |
Affects CDR-H2* |
| 73 |
3 |
Affects CDR-H1 |
| 76 |
3 |
Affects CDR-H1 |
| 78 |
3 |
Affects CDR-H1 |
| 94 |
4 |
Affects CDR-H3* |
| max |
57 |
|
[0166] After the residue positions are assigned a numerical score, all of the residue scores
are totaled. The acceptor germline sequence is the one with the highest total score.
In a case where two or more germline sequences have identical scores, then add 1 to
the total for each position where the non-human and human sequences are IDENTICAL
for the following FR residues: 1-23, 25, 36, 38-47, 66, 68, 70, 72, 74, 75, 77, and
79-93 (max 60). The residue scores are totaled again, and the acceptor germline sequence
is the one with the highest total score. If two or more germline sequences still have
identical scores, either one can be used as the acceptor germline sequence.
[0167] If the V
L sequence is a member of the kappa subclass of V
L, the non-human V
L sequence from the GITR specific antibody is compared to a group of four human V
L kappa germline amino acid sequences. The four sequences are comprised of one representative
from each of four established human V
L subgroups listed in
V. Barbie & M.-P. Lefranc, Exp. Clin. Immunogenetics 15:171-183 (1998) and
M.-P. Lefranc, Exp. Clin. Immunogenetics 18:161-174 (2001). The four sequences also correspond to the four subgroups listed in Kabat et al.,
U. S. Department of Health and Human Services, NIH Pub. 91-3242, pp. 103-130 (5th
Ed., 1991). The comparison of the non-human sequence to the four germline sequences
begins with the assignment of residue numbers to the non-human V
L sequence residues according to Kabat et al., U. S. Department of Health and Human
Services, NIH Pub. 91-3242 (5th Ed., 1991). The non-human V
L sequences are then aligned with each of the four human germline sequences. Since
the V genes only comprise V
L residues 1-95, only these residues are considered in the alignment. Next, the complementarity-determining
(CDR) and framework (FR) regions are delineated in the sequence. CDR and FR are delineated
according to the combination of the definitions provided in Kabat et al., U. S. Department
of Health and Human Services, NIH Pub. 91-3242 (5th Ed. 1991), and
C. Chothia & A.M. Lesk, J. Mol. Biol., 196:901-917 (1987). Therefore, the CDR definition used is residues 24-34 for CDR1, residues 50-56 for
CDR2, and residues 89-97 for CDR3 of the V
L domain. The next step involves assigning a numerical score at identified residue
position where the non-human and human sequences are identical. One example of this
scoring is shown in Table 5 below.
| Table 5 |
| |
Residue # |
Score |
Reason |
| |
2 |
4 |
Affects CDR-L1,3* |
| |
25 |
4 |
Affects CDR-L1* |
| |
29 |
4 |
Affects CDR-L1,3* |
| |
34 |
2 |
VL/VH interface |
| |
43 |
2 |
VL/VH interface |
| |
55 |
2 |
VL/VH interface |
| |
58 |
3 |
Affects CDR-L2 |
| |
89 |
2 |
VL/VH interface |
| |
91 |
2 |
VL/VH interface |
| |
94 |
2 |
VL/VH interface |
| |
max |
27 |
|
[0168] After the residue positions are assigned a numerical score, all of the residue scores
are totaled. The acceptor germline sequence is the one with the highest total score.
In a case where two or more germline sequences have identical scores, then add 1 to
the total for each position where the non-human and human sequences are IDENTICAL
for the following FR residues: 1-3, 5-23, 35-42, 44-49, 57, 59-88 (max 67). The residue
scores are totaled again, and the acceptor germline sequence is the one with the highest
total score. If two or more germline sequences still have identical scores, either
one can be used as the acceptor germline sequence.
[0169] The above parental monoclonal antibodies were humanized using this method. SEQ ID
NOs: 90, 92, 94, 96, 98, 100, 102, 104, 106, 108, and 110 are the sequences of variable
heavy chains polypeptides, and SEQ ID NOs: 91, 93, 95, 97, 99, 101, 103, 105, 107,
109, and 111 are the sequences of the variable light chains.
Example 3
Determining the Equilibrium Dissociation Constant (Kd) for Anti-human GITR Antibodies Using KinExA Technology
[0170] The equilibrium dissociation constants (K
d) for anti human GITR antibodies are determined using the KinExA 3000 instrument.
Sapidyne Instruments Inc., Boise Idaho, USA. KinExA uses the principle of the Kinetic
Exclusion Assay method based on measuring the concentration of uncomplexed antibody
in a mixture of antibody, antigen and antibody-antigen complex. The concentration
of free antibody is measured by exposing the mixture to a solid-phase immobilized
antigen for a very brief period of time. In practice, this is accomplished by flowing
the solution phase antigen-antibody mixture past antigen-coated particles trapped
in a flow cell. Data generated by the instrument are analyzed using custom software.
Equilibrium constants are calculated using a mathematical theory based on the following
assumptions:
- 1. The binding follows the reversible binding equation for equilibrium:

- 2. Antibody and antigen bind 1:1 and total antibody equals antigen-antibody complex
plus free antibody.
- 3. Instrument signal is linearly related to free antibody concentration.
[0171] PMMA particles (Sapidyne, Cat No. 440198) are coated with biotinylated GITR (or a
fragment thereof, such as the extracellular domain) according to Sapidyne "Protocol
for coating PMMA particles with biotinylated ligands having short or nonexistent linker
arms." EZ-link TFP PEO-biotin (Pierce, Cat. No. 21219) is used for biotinylation of
GITR, as per the manufacturer's recommendations (Pierce bulletin 0874).
Example 4
Determining the Equilibrium Dissociation Constant (Kd) for Humanized Anti-human GITR Antibodies Using BIAcore Technology
[0172] BIAcore determinations are performed essentially as described at Example 4 of copending,
commonly assigned
U.S. Patent Application No. 11/511,635 (filed 29-August-2006). Briefly, binding partners are immobilized on a BIAcore CM5 sensor chip using standard
amine-coupling procedure. Kinetic constants for the various interactions are determined
using BIAevaluation software 3.1. The K
d is determined using the calculated dissociation and association rate constants.
[0173] GITR antibodies 36E5, 3D6, 61G6, 6H6, 61F6,1D8, 17F10, 35D8, 49A1, 9E5, and 31H6
had the following Kd values:
Table 6: Affinity measurements of GITR antibodies
| Analyte (mAb) |
Capture antigen |
Ka (1/Ms) (x105) |
Kd (1/s) (x10-6) |
Apparent Kd (pM) |
| 36E5 |
hGITR-hIgG |
8.64 |
177 |
205 |
| 61F6 |
hGITR-hIgG |
11.1 |
13530 |
12189 |
| 61G6 |
hGITR-hIgG |
0.04 |
69 |
15602 |
| 3D6 |
hGITR-hIgG |
0.95 |
766 |
8046 |
| 6H6 |
hGITR-hIgG |
6.10 |
919 |
1507 |
| 1D8 |
hGITR-hIgG |
4.28 |
196 |
458 |
| 17F10 |
hGITR-hIgG |
13.1 |
146 |
111 |
| 35D8 |
hGITR-hIgG |
8.01 |
200 |
250 |
| 49A1 |
hGITR-hIgG |
4078 |
318 |
665 |
| 9E5 |
hGITR-hIgG |
23.3 |
27 |
12 |
| 31H6 |
hGITR-hIgG |
15.7 |
7 |
≤ 5 |
Example 5
Bioassays for the Assessment of Activating Anti-GITR Antibodies
[0174] The ability of a monoclonal antibody to biologically enhance GITR activity was assessed
by the effect on proliferation of naive T cells (see, e.g.,
Ito et al., (2006) PNAS 103(35):13138-43. Naive CD4
+ T cells were isolated from peripheral blood mononuclear cells (PBMC) by Ficoll centrifugation
followed by using a naive CD4 T cell isolation kit from STEMCELL Technologies.
[0175] In 96-well tissue flat-bottom culture plates, a total of 2x10
4 freshly purified naive CD4 T cells were co-cultured with irradiated CD32-expressing
L cells in the presence of an anti-hGITR antibody or the isotype control, which had
been pre-coated with anti-CD3.
[0176] Tables 7A and 7B shows the effect of varying doses of anti-GITR antibodies on proliferation
of naive CD4+ T cells (KM4-R63 is an isotype control antibody).
Table 7A
| Ab conc (ng/mL ) |
36E5.A5 |
JL5.3D6 |
61G6.B6 |
6H6.C3 |
61F6.B9 |
KM4.R63 |
| mean |
SD |
mean |
SD |
mean |
SD |
mean |
SD |
mean |
SD |
mean |
SD |
| 10000 |
83784 |
5854 |
96205 |
9562 |
135047 |
7362 |
91873 |
4218 |
93373 |
4099 |
29662 |
7817 |
| 2500 |
71843 |
7556 |
109291 |
21713 |
115725 |
9792 |
107347 |
6049 |
96296 |
1233 |
27007 |
4572 |
| 625 |
82075 |
6760 |
111455 |
5596 |
75125 |
5258 |
120374 |
10489 |
105194 |
5043 |
25503 |
4699 |
| 156 |
97139 |
11937 |
108929 |
5934 |
45588 |
6309 |
122653 |
7164 |
107643 |
8700 |
27496 |
4019 |
| 39.1 |
90331 |
6422 |
124377 |
26014 |
40075 |
2611 |
103621 |
2111 |
107473 |
2179 |
25650 |
197 |
| 9.8 |
78668 |
11867 |
79317 |
2260 |
34335 |
3038 |
59965 |
4439 |
96267 |
10551 |
24722 |
3890 |
| 2.4 |
47467 |
9197 |
43088 |
1264 |
28872 |
2754 |
35629 |
3144 |
61257 |
3294 |
24851 |
1945 |
| 0.61 |
31043 |
6389 |
33847 |
1985 |
32642 |
3155 |
30245 |
12806 |
37587 |
1858 |
29232 |
7135 |
| 0.15 |
26240 |
8161 |
30774 |
7303 |
35642 |
4447 |
33443 |
8983 |
31955 |
2689 |
26627 |
1010 |
| 0.04 |
27502 |
4280 |
33342 |
3656 |
30969 |
1537 |
38543 |
8259 |
34931 |
5787 |
31039 |
3042 |
Table 7B
| Ab conc (ng/mL) |
1D8.B5 |
35D8.B10 |
49A1.B1 |
9E5.C1 |
31H6.B7 |
17F10.B1 |
| mean |
SD |
mean |
SD |
mean |
SD |
mean |
SD |
mean |
SD |
mean |
SD |
| 1000.0 |
-- |
-- |
97278 |
3127 |
78031 |
5394 |
83553 |
6596 |
87531 |
1051 |
89358 |
12570 |
| 416.7 |
-- |
-- |
89761 |
9163 |
86306 |
3807 |
79685 |
7730 |
86915 |
2652 |
85702 |
9724 |
| 173.6 |
90962 |
3417 |
96241 |
3423 |
93594 |
4229 |
84579 |
5929 |
96935 |
4442 |
87519 |
7556 |
| 72.3 |
102810 |
3353 |
92371 |
5048 |
96126 |
5395 |
85291 |
16030 |
98595 |
7374 |
83511 |
5009 |
| 30.1 |
112003 |
5405 |
95258 |
8152 |
95517 |
6187 |
83407 |
5503 |
94683 |
7610 |
86986 |
2717 |
| 12.6 |
115163 |
6429 |
86232 |
5329 |
86001 |
1893 |
85659 |
5087 |
90531 |
3957 |
90231 |
2079 |
| 5.2 |
98161 |
5423 |
71405 |
10471 |
72192 |
1776 |
77653 |
5524 |
79605 |
7438 |
77786 |
5065 |
| 2.2 |
78492 |
1831 |
59817 |
745 |
61053 |
239 |
69187 |
7450 |
67645 |
1972 |
73420 |
12592 |
| 0.9 |
64788 |
773 |
50713 |
2257 |
54096 |
2816 |
60922 |
7139 |
56972 |
4470 |
61950 |
4598 |
| 0.4 |
60794 |
5069 |
52287 |
5015 |
54348 |
1018 |
60105 |
6687 |
58490 |
582 |
58704 |
7820 |
Example 5
Treatment of Tumors with TGF-β and GITR Antibodies
[0177] Previous studies revealed that gene expression of 4T1 tumors had increased levels
of TGF-β mRNA. It was hypothesized that immune co-stimulation by anti-GITR agonist
combined with removal of immune suppression by inhibition of TGF-β signaling would
induce synergistic anti-tumor efficacy.
[0178] To test this hypothesis, 1.5 x 10
5 4T1 tumor cells were implanted subcutaneously at the right flank of Balb/C mice.
Four or seven days after tumor implantation, a neutralization antibody to murine TGF-β
(1D11; Bioexpress) was injected at 100 µgs/200 µL subcutaneously at the neck and repeated
every three days for a total of 7 doses. Anti-mouse GITR agonist antibody, DTA-1,
was injected at day 7, 14, and 21 at 500 µg/200 µL. Tumor volume was measured every
three days. As shown in Table 8 below, DTA-1 or anti-TGF-β alone has little effect
by themselves. Combined treatment induces synergistic effect on either of the starting
days of anti-TGF-β treatment (day 4 or day 7 post tumor implantation). Values are
tumor volume (mm
3).
Table 8: Anti-tumor efficacy by anti-mGITR and/or anti-TGF-β
| Post tumor Imp |
IgG2b |
DTA1 |
anti-TG-β(D7) |
DTA1 + anti-TGF-β(D7) |
anti-TGF-β(D4) |
DTA1 + anti-TGF-β(D4) |
| MEAN |
SD |
N |
MEAN |
SD |
N |
MEAN |
SD |
N |
MEAN |
SD |
N |
MEAN |
SD |
N |
MEAN |
SD |
N |
| Day 7 |
57.476 |
4.383 |
12 |
55.4614 |
5.28 |
12 |
55.7903 |
6.85 |
12 |
49.1208 |
4.24 |
12 |
54.551 |
6.01 |
10 |
48.432 |
6.80 |
10 |
| Day 11 |
160.904 |
11.87 |
12 |
131.563 |
10.67 |
12 |
141.827 |
14.94 |
12 |
67.9306 |
5.98 |
12 |
140.118 |
15.54 |
10 |
70.202 |
3.65 |
10 |
| Day 14 |
259.459 |
10.42 |
12 |
213.981 |
20.49 |
12 |
222.383 |
18.47 |
12 |
58.5907 |
5.01 |
12 |
193.78 |
26.52 |
10 |
39.651 |
9.522 |
10 |
| Day 18 |
408.553 |
24.34 |
12 |
340.876 |
21.30 |
12 |
309.058 |
24.45 |
12 |
100.977 |
14.92 |
12 |
315.661 |
42.47 |
10 |
45.928 |
20.59 |
10 |
| Day 21 |
627.802 |
34.23 |
12 |
519.613 |
29.11 |
12 |
524.622 |
44.18 |
12 |
185.615 |
16.66 |
12 |
515.88 |
55.56 |
10 |
92.660 |
45.67 |
10 |
| Day 25 |
810.945 |
41.97 |
12 |
761.875 |
45.07 |
12 |
699.235 |
47.79 |
12 |
336.241 |
27.26 |
12 |
718.21 |
73.21 |
10 |
213.447 |
63.72 |
10 |
| Day 29 |
1099.43 |
33.47 |
12 |
1006.18 |
39.89 |
12 |
908.552 |
32.36 |
12 |
511.284 |
32.98 |
12 |
956.36 |
83.12 |
10 |
382.504 |
94.91 |
10 |
Example 6
Antibody-Radiation Combined Treatment of CT26 Tumors
[0179] CT26 tumor cells (3 x 10
5) were implanted subcutaneously on left flanks of Balb/c mice. Local irradiation (10
Gy) was applied to tumors that grew to 300 mm
3, after observing that DTA-1 alone did not have tumor-killing efficacy. A day after
the irradiation, DTA-1 (500ug) was injected subcutaneously on the neck area and repeated
every week for a total of three doses. Tumor volume was measured every two to five
days. In the group of 10 mice that underwent local irradiation and DTA-1 combined
treatment, 5 mice completely rejected the tumors and survived up to 3 months. DTA-1
or irradiation alone did not exhibit tumor rejection (see, e.g., Figure 1).
Example 7
Epitope Mapping of GITR antibodies
[0180] As noted above, DTA-1 is an agonist antibody raised again mouse GITR (see, e.g.,
Shimizu, et al.
supra). DTA-1 has been shown to have potent anti-tumor activities in mouse models of cancer
(see, e.g.,
Cohen, et al. (2006) Cancer Res. 66:4904-4912;
Ramirez-Montagut, et al. (2006) J. Immunol. 176:6434-6442;
Zhou, et al. (2007) J. Immunol. 179:7365-7375; and
Ko, et al. (2005) J. Exp. Med. 202:885-891).
[0181] To determine if the antibodies described above bound to a DTA-1-like epitope on the
human GITR protein, the DTA-1 epitope was first mapped on the mouse GITR protein.
Without a crystal structure of human or mouse GITR available, the DTA-1 epitope was
determined using standard site directed mutagenesis techniques (see, e.g.,
Kunkel (1985) Proc. Natl. Acad. Sci. 82:488-492) and the general principles of modularity of the TNF-receptor family (see, e.g.,
Naismith and Sprang,
supra).
[0182] Once the mouse GITR epitope recognized by DTA-1 was determined, the corresponding
residues on human GITR were changed to the mouse residues thereby conferring DTA-1
binding to human GITR. From this, it was determined the DTA-1-like epitope on human
GITR spanned modules 3 and 4 (see Figure 2), and the human GITR (SEQ ID NO: 89) epitope
recognized by two of the above-identified antibodies comprised Gly
57, Arg
65, His
67, Lys
80, Phe
81, Ser
82, and Gln
86.
Example 8
Treatment of Viral Infections with Anti-GITR Antibodies
[0183] HIV infection is characterized by defects in the generation and maintenance of central
memory cells. CD8+ central memory cells have a shorter half-life and are less abundant
in HIV-infected individuals than in controls. Also, the frequency of both CD4+ and
CD8+ HIV-specific T cells decreases rapidly after initiation of highly active antiretroviral
therapy (HAART). Co-stimulation on CD4+ by anti-GITR may provide a mechanism to increase
memory CD8+ response and to contribute to clearance of the virus. It has been shown
that treatment of persistently Friend virus-infected mice with anti-GITR antibody
to ameliorate suppression by Tregs significantly improved IFN-γ production by the
CD8+ T cells and allowed a significant reduction in viral loads (
Dittmer et al., (2004) Immunity 20: 293-303).
[0184] Another characteristic of HIV infection is massive apoptosis of CD4+ T cells starting
early in HIV infection. The progressive apoptotic deletion of CD4 T cells contributes
to weakened HIV-specific cellular immune responses and to the development of AIDS.
GITR co-stimulation has been shown to enhance murine antigen-specific cytokine secretion
by protecting T cells from apoptosis.
Lahey et al. (2007) J Infect Dis. 196: 43-49) demonstrated that anti-GITR treatment of HIV-specific CD4+ T cells enhances their
cytokine expression and protects them from apoptosis.
[0185] For infections resulting from viral causes, the antibodies of the invention may be
combined by application simulatenous with, prior to or subsequent to application of
standard therapies for treating viral infections. Such standard therapies vary depending
upon type of virus, although in almost all cases, administration of human serum containing
antitibodies (e.g., IgA, IgG) specific to the virus can be effective.
[0186] Influenza infection results in fever, cough, myalgia, headache and malaise, which
often occur in seasonal epidemics. Influenza is also associated with a number of postinfectious
disorders, such as encephalitis, myopericarditis, Goodpasture's syndrome, and Reye's
syndrome. Influenza infection also suppresses normal pulmonary antibacterial defenses,
such that patient's recovering from influenza have an increased risk of developing
bacterial pneumonia.
[0187] Influenza viral surface proteins show marked antigenic variation, resulting from
mutation and recombination. Thus, cytolytic T lymphocytes are the host's primary vehicle
for the elimination of virus after infection. Influenza is classified into three primary
types: A, B and C. Influenza A is unique in that it infects both humans and many other
animals (e.g., pigs, horses, birds and seals) and is the principal cause of pandemic
influenza. Also, when a cell is infected by two different influenza A strains, the
segmented RNA genomes of two two parental virus types mix during replication to create
a hybrid replicant, resulting in new epidemic strains. Influenza B does not replicate
in animals and thus has less genetic variation and influenza C has only a single serotype.
[0188] Most conventional therapies are palliatives of the symptoms resulting from infection,
while the host's immune response actually clears the disease. However, certain strains
(e.g., influenza A) can cause more serious illness and death. Influenza A may be treated
both clinically and prophylactically by the administration of the cyclic amines inhibitors
amantadine and rimantadine, which inhibit viral replication. However, the clinical
utility of these drugs is limited due to the relatively high incidence of adverse
reactions, their narrow anti-viral spectrum (influenza A only), and the propensity
of the virus to become resistant. The administration of serum IgG antibody to the
major influenza surface proteins, hemagglutinin and neuraminidase can prevent pulmonary
infection, whereas mucosal IgA is required to prevent infection of the upper respiratory
tract and trachea. The most effective current treatment for influenza is vaccination
with the administration of virus inactivated with formalin or β-propiolactone.
[0189] After an incubation of 9-11 days, hosts infected with the measles virus develope
fever, cough, coryza and conjunctivitis. Within 1-2 days, an erythematous, maculopapular
rash develop, which quickly spreads over the entire body. Because infection also suppresses
cellular immunity, the host is at greater risk for developing bacterial superinfections,
including otitis media, pneumonia and postinfectious encephalomyelitis. Acute infection
is associated with significant morbidity and mortality, especially in malnourished
adolescents.
[0190] Treatment for measles includes the passive administration of pooled human IgG, which
can prevent infection in non-immune subjects, even if given up to one week after exposure.
[0191] However, prior immunization with live, attenuated virus is the most effective treatment
and prevents disease in more than 95% of those immunized. As there is one serotype
of this virus, a single immunization or infection typically results in protection
for life from subsequent infection.
[0192] In a small proportion of infected hosts, measles can develop into SSPE, which is
a chronic progressive neurologic disorder resulting from a persistent infection of
the central nervous system. SSPE is caused by clonal variants of measles virus with
defects that interfere with virion assembly and budding. For these patients, reactivation
of T-cells with the antibodies of the invention so as to facilitate viral clearance
would be desirable.
[0193] Hepatitis B virus (HB-V) is the most infectious known bloodborne pathogen. It is
a major cause of acute and chronic heptatis and hepatic carcinoma, as well as life-long,
chronic infection. Following infection, the virus replicates in hepatocytes, which
also then shed the surface antigen HBsAg. The detection of excessive levels of HBsAg
in serum is used a standard method for diagnosing a hepatitis B infection. An acute
infection may resolve or it can develop into a chronic persistent infection.
[0194] Current treatments for chronic HBV include α-inteferon, which increases the expression
of class I human leukocyte antigen (HLA) on the surface of hepatocytes, thereby facilitating
their recognition by cytotoxic T lymphocytes. Additionally, the nucleoside analogs
ganciclovir, famciclovir and lamivudine have also shown some efficacy in the treatment
of HBV infection in in clinical trial. Additional treatments for HBV include pegylated
α-interferon, adenfovir, entecavir and telbivudine. While passive immunity can be
conferred through parental administration of anti-HBsAg serum antibodies, vaccination
with inactivated or recombinant HBsAg also confers resistance to infection. The anti-GITR
antibodies of the invention may be combined with conventional treatments for hepatitis
B infections for therapeutic advantage.
[0195] Hepatitis C virus (HC-V) infection may lead to a chronic form of hepatitis, resulting
in cirrosis. While symptoms are similar to infections resulting from Hepatitis B,
in distinct contrast to HB-V, infected hosts can be asymptomatic for 10-20 years.
Treatment for HC-V infection includes the administration of a combination of α-interferon
and ribavirin. A promising potential therapy for HC-V infection is the protease inhibitor
telaprevir (VX-960). Additional treatments include: anti-PD-1 antibody (MDX-1106,
Medarex), bavituximab (an antibody that binds anionic phospholipid phosphatidylserine
in a B2-glycoprotein I dependent manner, Peregrine Pharmaceuticals), anti-HPV viral
coat protein E2 antibod(y)(ies) (E.g., ATL 6865-Ab68+Ab65, XTL Pharmaceuticals) and
Civacir® (polyclonal anti-HCV human immune globulin). The anti-GITR antibodies of
the invention may be combined with one or more of these treatments for hepatitis C
infections for therapeutic advantage.
1. An antibody or antigen-binding fragment thereof that binds to human GITR comprising:
CDR-L1, CDR-L2 and CDR-L3 of the light chain variable domain comprising the amino
acid sequence set forth in SEQ ID NO: 105; and
CDR-H1, CDR-H2 and CDR-H3 of the heavy chain variable domain comprising the amino
acid sequence set forth in SEQ ID NO: 104.
2. An antibody or antigen-binding fragment thereof that binds to human GITR comprising
a light chain variable domain comprising an amino acid sequence which is at least
95% identical to the amino acid sequence set forth in SEQ ID NO: 105; and a heavy
chain variable domain comprising an amino acid sequence which is at least 95% identical
to the amino acid sequence set forth in SEQ ID NO: 104.
3. The antibody or antigen-binding fragment of claim 1 or 2, the light chain variable
domain of which has an asparagine or a glutamine residue at the position corresponding
to position 31 of the amino acid sequence set forth in SEQ ID NO: 105.
3. The antibody or antigen-binding fragment of claim 1 or 2, the light chain variable
domain of which has an asparagine or a glutamine residue at the position corresponding
to position 57 of the amino acid sequence set forth in SEQ ID NO: 105.
5. An antibody or antigen-binding fragment thereof that binds to human GITR comprising:
a CDR-L1 that comprises the amino acid sequence set forth in SEQ ID NO: 56 or a variant
thereof,
wherein the variant comprises one conservative amino acid substitution relative to
SEQ ID NO: 56;
a CDR-L2 that comprises the amino acid sequence set forth in SEQ ID NO: 67 or variant
thereof,
wherein the variant comprises one conservative amino acid substitution relative to
SEQ ID NO: 67;
a CDR-L3 that comprises the amino acid sequence set forth in SEQ ID NO: 78 or a variant
thereof,
wherein the variant comprises one conservative amino acid substitution relative to
SEQ ID NO: 78;
a CDR-H1 that comprises the amino acid sequence set forth in SEQ ID NO: 23 or a variant
thereof,
wherein the variant comprises one conservative amino acid substitution relative to
SEQ ID NO: 23;
CDR-H2 that comprises the amino acid sequence set forth in SEQ ID NO: 34 or a variant
thereof,
wherein the variant comprises one conservative amino acid substitution relative to
SEQ ID NO: 34; and
CDR-H3 that comprises the amino acid sequence set forth in SEQ ID NO: 45 or a variant
thereof,
wherein the variant comprises one conservative amino acid substitution relative to
SEQ ID NO: 45.
6. The antibody or antigen-binding fragment of any one of claims 1-5, which is an antigen-binding
fragment selected from the group consisting of Fab, Fab', Fab'-SH, Fv, scFv, F(ab')2, and a diabody.
7. An antibody or antigen-binding fragment thereof, produced by the hybridoma PTA-9890
deposited at the American Type Culture Collection (ATCC).
8. An antibody or antigen-binding thereof that binds to human GITR, wherein the antibody
or antigen-binding fragment recognizes an epitope spanning module 3 and module 4 of
human GITR protein (SEQ ID NO: 89).
9. The antibody or antigen-binding fragment of claim 8, wherein the epitope comprises
Gly57, Arg65, His67, Lys80, Phe81, Ser82, and Gln86.
10. An antibody or antigen-binding fragment thereof that is able to block binding of
the antibody or antigen-binding fragment of any one of claims 1-9 to human GITR in
a cross-blocking assay.
11. The antibody or antigen-binding fragment of any one of claims 1-10 which is an antibody.
12. The antibody or antigen-binding fragment of any one of claims 1-10 which is a monoclonal
antibody.
13. The antibody or antigen-binding fragment of any one of claims 1-10 which is a humanized
antibody.
14. The antibody or antigen-binding fragment of any one of claims 1-13 wherein the heavy
chain is fused to a γ1 human heavy chain constant region.
15. The antibody or antigen-binding fragment of any one of claims 1-14, comprising a
γ1 human heavy chain constant region or a variant thereof, wherein the constant region
variant comprises up to 20 conservatively modified amino acid substitutions.
16. The antibody or antigen-binding fragment of any one of claims 1-14, comprising a
γ4 human heavy chain constant region or a variant thereof, wherein the constant region
variant comprises up to 20 conservatively modified amino acid substitutions.
17. A pharmaceutical composition comprising the antibody or antigen-binding of any one
of claims 1-16 and a pharmaceutically acceptable carrier.
18. A nucleic acid encoding the light chain variable domain and/or the heavy chain variable
domain of the antibody or antigen-binding fragment of any one of claims 1-16.
19. An expression vector comprising the nucleic acid of claim 18 operably linked to expression
control sequences that are recognized by a host cell when the host cell is transfected
with the vector.
20. A host cell comprising the expression vector of claim 19.
21. The host cell of claim 20 which is a Chinese hamster ovary cell or a Pichia pastoris cell.
22. A method of producing an antibody or antigen-binding fragment thereof that binds
to human GITR comprising: culturing the host cell of claim 20 or 21 in culture medium
under conditions wherein the nucleic acid sequence is expressed, thereby producing
the antibody or antigen-binding fragment thereof comprising the encoded light and
heavy chain variable domains; and recovering the antibody or antigen-binding fragment
thereof from the host cell or culture medium.
23. The antibody or antigen-binding fragment thereof that binds to human GITR, which
is obtainable by the method of claim 22.
24. An antibody or antigen-binding fragment thereof of any one or claims 1-16 or 23 for
use in a method of treatment of a human subject by therapy.
25. The antibody or antigen-binding fragment thereof for use of claim 24, wherein the
therapy is by enhancing an immune response in a human subject.
26. The antibody or antigen-binding fragment thereof for use of claim 24 or 25, wherein
the antibody or antigen-binding fragment thereof is co-administered with a second
therapeutic agent.
27. The antibody or antigen-binding fragment thereof for use of claim 26, wherein the
second therapeutic agent is an antibody.
28. The antibody or antigen-binding fragment thereof for use of claim 26, wherein the
second therapeutic agent is an anti-TGFβ antibody.
29. The antibody or antigen-binding fragment thereof for use of claim 24 or 25, wherein
the antibody or antigen-binding fragment is co-administered with local radiation.
30. The antibody or antigen-binding fragment thereof for use of claim 26, wherein the
second therapeutic agent is selected from the group consisting of thiotepa, busulfan,
improsulfan, piposulfan, benzodopa, carboquone, meturedopa, uredopa, altretamine,
triethylenemelamine, trietylenephosphoramide, triethiylenethiophosphoramide, trimethylolomelamine,
bullatacin, bullatacinone, camptothecin, topotecan, bryostatin, callystatin, CC-1065,
cryptophycin 1, cryptophycin 8, dolastatin, duocarmycin, KW-2189, CB 1-TM1, eleutherobin,
pancratistatin, sarcodictyin, spongistatin, chlorambucil, chlomaphazine, cholophosphamide,
estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan,
novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard, carmustine,
chlorozotocin, fotemustine, lomustine, nimustine, ranimnustine, calicheamicin, calicheamicin
gamma II, calicheamicin omega II, dynemicin, dynemicin A, clodronate, neocarzinostatin
chromophore, aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin,
carabicin, caminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin,
detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin, morpholino-doxorubicin, cyanomorpholino-doxorubicin,
2-pyrrolino-doxorubicin, deoxydoxorubicin, epirubicin, esorubicin, idarubicin, marcellomycin,
mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin,
puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex,
zinostatin, zorubicin, 5-fluorouracil (5-FU), denopterin, methotrexate, pteropterin,
trimetrexate, fludarabine, 6-mercaptopurine, thiamiprine, thioguanine, ancitabine,
azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine,
floxuridine, calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone,
aminoglutethimide, mitotane, trilostane, frolinic acid, aceglatone, aldophosphamide
glycoside, aminolevulinic acid, eniluracil, amsacrine, bestrabucil, bisantrene, edatraxate,
defofamine, demecolcine, diaziquone, elformithine, elliptinium acetate, epothilone,
etoglucid, gallium nitrate, hydroxyurea, lentinan, lonidainine, maytansine, ansamitocins,
mitoguazone, mitoxantrone, mopidanmol, nitraerine, pentostatin, phenamet, pirarubicin,
losoxantrone, podophyllinic acid, 2-ethylhydrazide, procarbazine, razoxane, rhizoxin,
sizofuran, spirogermanium, tenuazonic acid, triaziquone, 2,2',2"-trichlorotriethylamine,
T-2 toxin, verracurin A, roridin A, anguidine, urethane, vindesine, dacarbazine, mannomustine,
mitobronitol, mitolactol, pipobroman, gacytosine, arabinoside, cyclophosphamide, paclitaxel,
a cremophor-free, albumin-engineered nanoparticle formulation of paclitaxel, docetaxel,
chloranbucil, gemcitabine, 6-thioguanine, mercaptopurine, methotrexate, cisplatin,
carboplatin, vinblastine, platinum, etoposide, ifosfamide, mitoxantrone, vincristine,
vinorelbine, novantrone, teniposide, edatrexate, daunomycin, aminopterin, capecitabine,
ibandronate, CPT-11, RFS2000, difluoromethylornithine (DMFO), retinoic acid, tamoxifen,
raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone,
toremifene, 4(5)-imidazoles, aminoglutethimide, megestrol acetate, exemestane, formestanie,
fadrozole, vorozole, letrozole, anastrozole, flutamide, nilutamide, bicalutamide,
leuprolide, goserelin, troxacitabine, angiozyme, allovectin, leuvectin, vaxid, recombinant
IL-2, lurtotecan and abarelix.
31. The antibody or antigen-binding fragment thereof for use of claim 26, wherein the
second therapeutic agent is selected from the group consisting of an alkylating agent,
an ethylenimine, a methylamelamine, an acetogenin, a camptothecin, a cryptophycin,
a sarcodictyin, an antibiotic, a bisphosphonate, a mitomycin, an anti-metabolite,
a folic acid analogue, a purine analog, an androgen, an anti-adrenal, a folic acid
replenisher, an epothilone, a trichothecene, a taxoid, a retinoid, an anti-hormonal
agent, an aromatase inhibitor, an anti-androgen, an antisense oligonucleotide, a ribozyme,
a HER2 expression inhibitor and a vaccine.
32. The antibody or antigen-binding fragment thereof for use of claim 25, wherein the
immune response is against a proliferative disorder.
33. The antibody or antigen-binding fragment thereof for use of claim 25, wherein the
immune response is against cancer.
34. The antibody or antigen-binding fragment thereof for use of claim 33, wherein the
cancer is selected from the group consisting of adenocarcinoma, lymphoma, blastoma,
sarcoma, leukemia, squamous cell cancer, small-cell lung cancer, non-small cell lung
cancer, gastrointestinal cancer, Hodgkin's lymphoma, non-Hodgkin's lymphoma, pancreatic
cancer, glioblastoma, glioma, cervical cancer, ovarian cancer, liver cancer, bladder
cancer, breast cancer, colon cancer, colorectal cancer, endometrial carcinoma, myeloma,
salivary gland carcinoma, kidney cancer, basal cell carcinoma, melanoma, prostate
cancer, vulval cancer, thyroid cancer, testicular cancer, esophageal cancer, and head
and neck cancer.
35. The antibody or antigen-binding fragment thereof for use of claim 25, wherein the
immune response is against a viral infection.
36. The antibody or antigen-binding fragment thereof for use of claim 35, wherein the
viral infection is caused by a virus which is selected from the group consisting of
human papilloma viruses (HPV), herpes simplex virus, hepatitis B virus, hepatitis
C virus, measles virus, human immunodeficiency virus (HIV) and Epstein Barr virus
(EBV).